Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Anesth ; 29(1): 65-77, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24943455

RESUMEN

PURPOSE: The N- and C-terminal regions of dynorphin (Dyn) A (1-17) activate opioid and N-methyl-D-aspartate receptors, respectively. Earlier studies demonstrated that Dyn-converting enzyme cleaved Dyn A (1-17) mainly at the Arg(6)-Arg(7) bond, resulting in the production of N- and C-terminal region peptide fragments, and that this enzyme was not inhibited by a mixture of the three peptidase inhibitors (PIs) amastatin (A), captopril (C), and phosphoramidon (P). The purpose of the present study was to evaluate antinociceptive potential and toxicity with intracerebroventricular administration of Dyn A (1-17) or (1-13) under pretreatment with a mixture of A, C, and P and/or Dyn-converting enzyme inhibitor (p-hydroxymercuribenzoate). METHODS: Peptide fragments from Dyn A (1-17) following incubation with membrane preparation under pretreatment with a mixture of the three PIs was identified by matrix-assisted laser desorption ionization time-of-flight mass spectrometer (MALDI-TOF-MS). Infusion of drugs and peptides into the third ventricle in rats was performed via indwelling cannulae. Induction of antinociception and toxicity by Dyn A (1-17), Dyn A (1-13), Dyn A (1-6), or Dyn A (7-17) were determined by the tail-flick test and induction of barrel rotation, respectively. The effects of the PIs on antinociception and toxicity were evaluated by a dose-response study and a comparison of differences among various combinations of Dyn A (1-17) or Dyn A (1-13) and the three PIs and p-hydroxymercuribenzoate. RESULTS: MALDI-TOF-MS analysis identified Dyn A (1-6) and Dyn A (1-10) fragments as products following incubation of Dyn A (1-17) with membrane preparation of rat midbrain under pretreatment with a mixture of the three PIs. Pretreatment with a mixture of the three PIs produced an approximately 30-fold augmentation in antinociception induced by low-dose intracerebroventricular administration of Dyn A (1-17) or (1-13) in a µ-, δ- and κ-opioid receptor antagonist-reversible manner, but without signs of toxicity such as barrel rotation in the rat. Dyn A (1-17)-induced antinociception and toxicity was greater than that of Dyn A (1-6), Dyn A (1-13), or Dyn A (7-17) at the same dose. Dyn A (1-17)-induced antinociception and toxicity under pretreatment with various combinations of the three PIs and p-hydroxymercuribenzoate was greater than that with a mixture of the three PIs alone. CONCLUSION: These findings suggest that administration of a mixture of the three PIs increases Dyn A (1-17)- or (1-13)-induced antinociception under physiological conditions without toxicity.


Asunto(s)
Analgésicos Opioides/toxicidad , Analgésicos/efectos adversos , Analgésicos/farmacología , Dinorfinas/toxicidad , Inhibidores de Proteasas/farmacología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Animales , Química Encefálica/efectos de los fármacos , Captopril/administración & dosificación , Captopril/farmacología , Relación Dosis-Respuesta a Droga , Dinorfinas/administración & dosificación , Dinorfinas/farmacología , Glicopéptidos/administración & dosificación , Glicopéptidos/farmacología , Inyecciones Intraventriculares , Masculino , Dimensión del Dolor/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/farmacología , Ratas , Ratas Wistar , Receptores Opioides/efectos de los fármacos
2.
Acta Anaesthesiol Scand ; 53(1): 55-60, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19032555

RESUMEN

BACKGROUND: It has been proposed that the volatile anesthetic isoflurane induces neuroprotection and that the endogenous opioid peptide dynorphin induces neurocytotoxicity in cells. The levels of dynorphin are often significantly elevated in neuropathophysiological conditions, and dynorphin can directly induce toxicity. However, the neuroprotective effects of isoflurane on dynorphin-induced cytotoxicity are still unclear. METHODS: In order to determine the effect of isoflurane on dynorphin-induced cytotoxicity in neuronal cells, we have designed a device wherein cultured human neuroblastoma SH-SY5Y cells can be exposed to isoflurane. Fully differentiated SH-SY5Y cells were obtained by treating the cells with retinoic acid for 6 days. We examined SH-SY5Y cell survival, apoptosis, and antiapoptotic protein expression by cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling stain, and Western blot analysis, respectively. RESULTS: After 16 h of dynorphin (10 microM) treatment, the SH-SY5Y cells showed significant cytotoxicity, apoptosis, and downregulation of the antiapoptotic Bcl-2 protein expression. These effects of dynorphin were significantly inhibited by isoflurane exposure for 32 h [pretreatment for 16 h and posttreatment (after dynorphin treatment) for 16 h]. CONCLUSION: Thus, our results suggest that isoflurane exerts neuroprotective effects in the case of dynorphin-induced pathophysiological disruption.


Asunto(s)
Diferenciación Celular , Regulación hacia Abajo/efectos de los fármacos , Dinorfinas/toxicidad , Isoflurano/farmacología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos
3.
Neuroscience ; 116(3): 807-16, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12573721

RESUMEN

Dynorphin A (1-17) is an endogenous opioid peptide that is antinociceptive at physiological concentrations, but in excess can elicit a number of pathological effects. Both kappa-opioid and N-methyl-D-aspartate receptor antagonists modulate dynorphin toxicity, suggesting that dynorphin is acting directly or indirectly through these receptor types. We found in spinal cord neurons that the neurotoxic effects of dynorphin A and several dynorphin-derived peptide fragments are largely mediated by N-methyl-D-aspartate receptors. Despite these findings, aspects of dynorphin A toxicity could not be accounted for by opioid or N-methyl-D-aspartate receptor mechanisms. To address this issue, neurons enriched in kappa-opioid, N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptors were isolated from embryonic day-15 mouse striata and the effects of extracellularly administered dynorphin A (1-17) and (13-17) on neuronal survival were examined in vitro. Unlike spinal cord neurons, N-methyl-D-aspartate receptors mature later than alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainate receptors in striatal neurons, thus providing a strategy to elucidate non-N-methyl-D-aspartate receptor-mediated mechanisms of toxicity. Time-lapse photography was used to repeatedly follow the same neurons before and during experimental treatments. Dynorphin A (1-17 or 13-17; 10 microM) caused significant neuronal losses after 48 to 72 hours versus untreated controls. Dynorphin A or A (13-17) toxicity was unaffected by the opioid receptor antagonist naloxone (10 microM) or by dizocilpine (10 microM). In contrast, the AMPA/kainate receptor antagonist 6-cyano-7-nitroquinoxaline- 2,3-dione (10 microM) significantly attenuated only dynorphin A (1-17)-induced neuronal losses and not that induced by dynorphin A (13-17). Dynorphin A (1-17) toxicity was accompanied by a proportional loss of R2 and R3 subunits of the AMPA receptor complex, but not non-N-methyl-D-aspartateR1, expressing neurons and was mimicked by the ampakine 1-(1,4-benzodioxan-6-ylcarbonyl)piperidine. Although it is unclear whether dynorphin A activates alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainate receptors directly or indirectly via glutamate release, our culture conditions do not support glutamate retention or accumulation. Our findings suggest that dynorphin A (1-17) can exert toxic effects on striatal neurons via an alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainate receptor mechanism.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Dinorfinas/toxicidad , Neuronas/efectos de los fármacos , Receptores AMPA/fisiología , Receptores de Ácido Kaínico/fisiología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Cuerpo Estriado/patología , Femenino , Ratones , Ratones Endogámicos ICR , Neuronas/patología , Embarazo
4.
Exp Cell Res ; 269(1): 54-63, 2001 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-11525639

RESUMEN

Dynorphin A, a prodynorphin-derived peptide, is able to induce neurological dysfunction and neuronal death. To study dynorphin cytotoxicity in vitro, prodynorphin-derived peptides were added into the culture medium of nonneuronal and neuronal cells or delivered into these cells by lipofection or electroporation. Cells were unaffected by extracellular exposure when peptides were added to the medium. In contrast, the number of viable cells was significantly reduced when dynorphin A or "big dynorphin," consisting of dynorphins A and B, was transfected into cells. Big dynorphin was more potent than dynorphin A, whereas dynorphin B; dynorphin B-29; [Arg(11,13)]-dynorphin A(-13)-Gly-NH-(CH(2))(5)-NH(2), a selective kappa-opioid receptor agonist; and poly-l-lysine, a basic peptide more positively charged than big dynorphin, failed to affect cell viability. The opioid antagonist naloxone did not prevent big dynorphin cytotoxicity. Thus, the toxic effects were structure selective but not mediated through opioid receptors. When big dynorphin was delivered into cells by lipofection, it became localized predominantly in the cytoplasm and not in the nuclei. Big dynorphin appeared to induce toxicity through an apoptotic mechanism that may involve synergistic interactions with the p53 tumor-suppressor protein. It is proposed that big dynorphin induces cell death by virtue of its net positive charge and clusters of basic amino acids that mimic (and thereby perhaps interfere with) basic domains involved in protein-protein interactions. These effects may be relevant for a pathophysiological role of dynorphins in the brain and spinal cord and for control of death of tumor cells, which express prodynorphin at high levels.


Asunto(s)
Apoptosis/fisiología , Citotoxinas/farmacología , Dinorfinas/toxicidad , Degeneración Nerviosa/metabolismo , Fragmentos de Péptidos/farmacología , Receptores Opioides/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Resinas de Intercambio de Catión/farmacocinética , Compartimento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiopatología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Dinorfinas/metabolismo , Encefalinas/metabolismo , Inmunohistoquímica , Lípidos/farmacocinética , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/fisiopatología , Precursores de Proteínas/metabolismo , Estructura Terciaria de Proteína/fisiología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Proteína p53 Supresora de Tumor/efectos de los fármacos
5.
Exp Neurol ; 168(1): 78-87, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11170722

RESUMEN

Dynorphin A [dynorphin A (1-17)] is an endogenous opioid peptide that is antinociceptive at physiological concentrations. Levels of dynorphin A increase markedly following spinal cord trauma and may contribute to secondary neurodegeneration. Both kappa opioid and N-methyl-d-aspartate (NMDA) receptor antagonists can modulate the effects of dynorphin, suggesting that dynorphin is acting through kappa opioid and/or NMDA receptor types. Despite these findings, few studies have critically examined the mechanisms of dynorphin A neurotoxicity at the cellular level. To better understand how dynorphin affects cell viability, structure-activity studies were performed examining the effects of dynorphin A and dynorphin A-derived peptide fragments on the survival of mouse spinal cord neurons coexpressing kappa opioid and NMDA receptors in vitro. Time-lapse photography was used to repeatedly follow the same neurons before and during experimental treatments. Dynorphin A caused significant neuronal losses that were dependent on concentration (> or = 1 microM) and duration of exposure. Moreover, exposure to an equimolar concentration of dynorphin A fragments (100 microM) also caused a significant loss of neurons. The rank order of toxicity was dynorphin A (1-17) > dynorphin A (1-13) congruent with dynorphin A (2-13) congruent with dynorphin A (13-17) (least toxic) > dynorphin A (1-5) ([Leu(5)]-enkephalin) or dynorphin A (1-11). Dynorphin A (1-5) or dynorphin A (1-11) did not cause neuronal losses even following 96 h of continuous exposure, while dynorphin A (3-13), dynorphin A (6-17), and dynorphin A (13-17) were neurotoxic. The NMDA receptor antagonist MK-801 (dizocilpine) (10 microM) significantly attenuated the neurotoxic effects of dynorphin A and/or dynorphin-derived fragments except dynorphin A (13-17), suggesting that the neurotoxic effects of dynorphin were largely mediated by NMDA receptors. Thus, toxicity resides in the carboxyl-terminal portion of dynorphin A and this minimally includes dynorphin A (3-13) and (13-17). Our findings suggest that dynorphin A and/or its metabolites may contribute significantly to neurodegeneration during spinal cord injury and that alterations in dynorphin A biosynthesis, metabolism, and/or degradation may be important in determining injury outcome.


Asunto(s)
Dinorfinas/toxicidad , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Receptores de N-Metil-D-Aspartato/fisiología , Receptores Opioides kappa/fisiología , Médula Espinal/citología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Dinorfinas/química , Antagonistas de Aminoácidos Excitadores/toxicidad , Feto , Cinética , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/fisiología , Fragmentos de Péptidos/química , Médula Espinal/embriología , Relación Estructura-Actividad
6.
J Chem Neuroanat ; 17(4): 183-97, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10697245

RESUMEN

It has recently been demonstrated that selective inhibition of both neuronal constitutive and inducible nitric oxide synthases (ncNOS and iNOS) is neuroprotective in a model of dynorphin (Dyn) A(1-17)-induced spinal cord injury. In the present study, various methods including the conversion of 3H-L-arginine to 3H-citrulline, immunohistochemistry and in situ hybridization are employed to determine the temporal profiles of the enzymatic activities, immunoreactivities, and mRNA expression for both ncNOS and iNOS after intrathecal injection of a neurotoxic dose (20 nmol) of Dyn A(1-17). The expression of ncNOS immunoreactivity and mRNA increased as early as 30 min after injection and persisted for 1-4 h. At 24-48 h, the number of ncNOS positive cells remained elevated while most neurons died. The cNOS enzymatic activity in the ventral spinal cord also significantly increased at 30 min 48 h, but no significant changes in the dorsal spinal cord were observed. However, iNOS mRNA expression increased later at 2 h, iNOS immunoreactivity and enzymatic activity increased later at 4 h and persisted for 24-48 h after injection of 20 nmol Dyn A(1-17). These results indicate that both ncNOS and iNOS are associated with Dyn-induced spinal cord injury, with ncNOS predominantly involved at an early stage and iNOS at a later stage.


Asunto(s)
Óxido Nítrico Sintasa/genética , Traumatismos de la Médula Espinal/enzimología , Médula Espinal/enzimología , Animales , Dinorfinas/toxicidad , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , ARN Mensajero/genética , Ratas , Ratas Wistar , Valores de Referencia , Médula Espinal/citología , Médula Espinal/patología , Traumatismos de la Médula Espinal/inducido químicamente , Traumatismos de la Médula Espinal/patología , Transcripción Genética
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 22(2): 134-8, 2000 Apr.
Artículo en Chino | MEDLINE | ID: mdl-12903514

RESUMEN

OBJECTIVE: To elucidate the effects of N-methyl-D-aspartate(NMDA) receptor and nitric oxide synthase (NOS) activity in dynorphin (Dyn)-induced spinal cord injury. METHODS: The NMDA receptor activity was measured by radio-ligand of 3H-MK801. The constitutive and inducible NOS (cNOS and iNOS) activities were assayed by 3H-arginine conversion. RESULTS: In ventral samples, both 3H-MK801 binding and cNOS activity increased at 0.5 h and persisted for 48 h while iNOS activity enhanced at 4 h after intratheacal injection (i.t.) Dyn A(1-17) at dose of 20 nmol/L. However, the 3H-MK801 binding activity reduced significantly from 4 h to 24 h and cNOS activity did not change at the same time in dorsal samples. 7-nitroindozol (7-NI) and aminoguanidine (AG) inhibited the effects of Dyn A(1-17) (20 nmol/L) on 3H-MK801 binding and NOS activities in ventral samples. N-nitro-L-arginine methyl ester (L-NAME) did not affect the elevation of Dyn A(1-17) on NOS activities but caused 3H-MK801 binding activity reduction in ventral samples. CONCLUSIONS: NMDA-NOS pathway might play important role in Dyn spinal neurotoxicity. NOS inhibitors and Dyn might produce cooperative down-regulation on the function of NMDA-NOS pathway in dorsal cord.


Asunto(s)
Dinorfinas/toxicidad , Óxido Nítrico Sintasa/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Óxido Nítrico Sintasa/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/inducido químicamente
8.
J Neurotrauma ; 16(1): 85-98, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9989468

RESUMEN

The pharmacological effects of nitric oxide synthase (NOS) inhibitors, NO donor, and NOS substrate on dynorphin(Dyn) A(1-17) spinal neurotoxicity were studied. Intrathecal (i.t.) pretreatment with both 7-nitroindazole 1 micromol, a selective neuronal constitutive NOS (ncNOS) inhibitor, and aminoguanidine 1 micromol, a selective inducible NOS (iNOS) inhibitor, 10 min prior to i.t. Dyn A(1-17) 20 nmol significantly ameliorated Dyn-induced neurological outcome. Both 7-nitroindazole and aminoguanidine significantly antagonized the increases of cNOS and iNOS activities measured by conversion of 3H-L-arginine to 3H-L-citrulline in the ventral spinal cord, and blocked the Dyn-induced increases of ncNOS-immunoreactivity in the ventral horn cells 4 h after i.t. Dyn A(1-17) 20 nmol. Pretreatment with Nomega-nitro-L-arginine methyl ester (L-NAME) 1 micromol, a cNOS inhibitor nonselective to both ncNOS and endothelial NOS (ecNOS), did not antagonize Dyn A(1-17) 20 nmol-induced permanent paraplegia but aggravated Dyn A(1-17) 10 nmol-induced transient paralysis and caused permanent paraplegia. Pretreatment with L-NAME 1 micromol 10 min before i.t. Dyn A(1-17) 1.25 and 2.5 nmol, which produced no significant motor dysfunction alone, induced transient paralysis in seven out of 12 and five out of seven rats, respectively. L-NAME 1 micromol plus Dyn A(1-17) 10 nmol induced ncNOS-immunoreactivity expression in ventral horn cells. Both low and high doses of aminoguanidine (0.2-30 micromol) did not affect spinal motor function, but high doses of L-NAME (5-20 micromol) induced dose-dependent hindlimb and tail paralysis associated with spinal cord injury in normal rats. Pretreatment with low-dose Spermine NONOate, a controlled NO releaser, 0.1 and 0.5 micromol 10 min before i.t. Dyn A(1-17) 20 nmol, significantly prevented Dyn spinal neurotoxicity, and high-dose Spermine NONOate 2 micromol i.t. per se induced transient and incomplete paraplegia. But pretreatment with L-Arg 10 micromol 10 min before Dyn A(1-17) 20 nmol produced only partial blockade of Dyn-induced paraplegia. These results demonstrated that relatively specific inhibition of ncNOS and iNOS block Dyn-induced increases in cNOS and iNOS activities and ncNOS-immunoreactivity in ventral spinal cord, but nonspecific inhibition of ncNOS and ecNOS aggravated Dyn spinal neurotoxicity. It suggested that both ncNOS and iNOS play an important role, but ecNOS might be beneficial in Dyn spinal neurotoxicity. Moderate production of NO (at vascular level) has an apparently neuroprotective effect, and overproduction of NO (at cellular level) induces neurotoxicity.


Asunto(s)
Dinorfinas/toxicidad , Óxido Nítrico/fisiología , Médula Espinal/efectos de los fármacos , Animales , Arginina/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Inmunohistoquímica , Indazoles/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , Óxidos de Nitrógeno , Ratas , Ratas Wistar , Espermina/análogos & derivados , Espermina/farmacología , Médula Espinal/enzimología , Médula Espinal/metabolismo , Factores de Tiempo
9.
Exp Neurol ; 160(2): 361-75, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10619553

RESUMEN

Dynorphin A is an endogenous opioid peptide that preferentially activates kappa-opioid receptors and is antinociceptive at physiological concentrations. Levels of dynorphin A and a major metabolite, dynorphin A (1-13), increase significantly following spinal cord trauma and reportedly contribute to neurodegeneration associated with secondary injury. Interestingly, both kappa-opioid and N-methyl-D-aspartate (NMDA) receptor antagonists can modulate dynorphin toxicity, suggesting that dynorphin is acting (directly or indirectly) through kappa-opioid and/or NMDA receptor types. Despite these findings, few studies have systematically explored dynorphin toxicity at the cellular level in defined populations of neurons coexpressing kappa-opioid and NMDA receptors. To address this question, we isolated populations of neurons enriched in both kappa-opioid and NMDA receptors from embryonic mouse spinal cord and examined the effects of dynorphin A (1-13) on intracellular calcium concentration ([Ca2+]i) and neuronal survival in vitro. Time-lapse photography was used to repeatedly follow the same neurons before and during experimental treatments. At micromolar concentrations, dynorphin A (1-13) elevated [Ca2+]i and caused a significant loss of neurons. The excitotoxic effects were prevented by MK-801 (Dizocilpine) (10 microM), 2-amino-5-phosphopentanoic acid (100 microM), or 7-chlorokynurenic acid (100 microM)--suggesting that dynorphin A (1-13) was acting (directly or indirectly) through NMDA receptors. In contrast, cotreatment with (-)-naloxone (3 microM), or the more selective kappa-opioid receptor antagonist nor-binaltorphimine (3 microM), exacerbated dynorphin A (1-13)-induced neuronal loss; however, cell losses were not enhanced by the inactive stereoisomer (+)-naloxone (3 microM). Neuronal losses were not seen with exposure to the opioid antagonists alone (10 microM). Thus, opioid receptor blockade significantly increased toxicity, but only in the presence of excitotoxic levels of dynorphin. This provided indirect evidence that dynorphin also stimulates kappa-opioid receptors and suggests that kappa receptor activation may be moderately neuroprotective in the presence of an excitotoxic insult. Our findings suggest that dynorphin A (1-13) can have paradoxical effects on neuronal viability through both opioid and non-opioid (glutamatergic) receptor-mediated actions. Therefore, dynorphin A potentially modulates secondary neurodegeneration in the spinal cord through complex interactions involving multiple receptors and signaling pathways.


Asunto(s)
Analgésicos Opioides/toxicidad , Dinorfinas/toxicidad , Neuronas/citología , Neurotoxinas/toxicidad , Fragmentos de Péptidos/toxicidad , Médula Espinal/citología , Animales , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Embrión de Mamíferos , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/farmacología , Ratones , Ratones Endogámicos ICR , Naloxona/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Valina/análogos & derivados , Valina/farmacología
10.
Yao Xue Xue Bao ; 32(2): 85-9, 1997.
Artículo en Chino | MEDLINE | ID: mdl-11243205

RESUMEN

Intrathecal(i.t.) injection of 10 microliters of dynorphin A(1-17) 20 nmol.L-1 per rat resulted in irreversible hind limb paralysis and suppression of the tail-flick reflex lasting for up to 40 h. The dual effects of dynorphin appeared 5-10 min after the i.t. administration. Histologic examination of the spinal cord in the rats demonstrated dead and/or dying and degenerated motor-neurons in the ventral horn located predominately in the lumbar segment(a 87.2% reduction of the number of motor neurons, P < 0.01) and also in a lesser degree in sacral segment(-69.6%, P < 0.05). The thoracic segment was essentially normal(-8.2%, P > 0.05).


Asunto(s)
Dinorfinas/toxicidad , Parálisis/inducido químicamente , Médula Espinal/efectos de los fármacos , Animales , Dinorfinas/administración & dosificación , Miembro Posterior , Inyecciones Espinales , Masculino , Neuronas Motoras/efectos de los fármacos , Ratas , Ratas Wistar
11.
Neurosci Lett ; 203(1): 13-6, 1996 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-8742035

RESUMEN

Nitric oxide (NO) mediation in the spinal cord injury induced by intrathecal (i.t.) dynorphin (Dyn) administration was studied with NADPH-diaphorase (Nd) histochemistry. Normally, there was rarely NO synthase (NOS) activity in spinal cord motomeurons, and Dyn A(1-17) 10 nmol, which produced only transient paralysis, did not induce Nd/NOS expression in ventral horn cells. After a paralyzing dose of i.t. Dyn A(1-17) 20 nmol, which definitely produced permanent paraplegia and neuronal death, Nd/NOS began to express in motoneurons at 30 min, increased in numbers and intensities at 2-4 h and persisted up to 8 h. Most of Nd/NOS motoneurons disappeared at 24 h coincident with the neuronal death. Quite a few intensively-stained Nd-positive small cells and swollen varicosities became visible only in rats with permanent paraplegia and neuronal death, beginning at 2 h, maximizing at 3-4 h and remaining up to 24 h. These results suggest that NOS expression was induced in the ventral horn of spinal cord, including small cells and varicosities as well as motoneurons closely correlated in time and degree with pathological changes in motoneurons caused by spinal Dyn neurotoxicity.


Asunto(s)
Células del Asta Anterior/efectos de los fármacos , Dinorfinas/toxicidad , Óxido Nítrico Sintasa/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Recuento de Células/efectos de los fármacos , Ratas , Ratas Wistar
12.
Pharmacol Biochem Behav ; 51(2-3): 387-90, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7667358

RESUMEN

Dynorphin A(1-13) blocks opiate withdrawal in rats without producing dependence, and enhances analgesia in morphine-tolerant animals. Its potential use in humans is therefore of interest. Dynorphin A(1-13) has little toxicity when administered at modest doses IV but has been reported to cause hindlimb paralysis and necrosis of the spinal cord in rats, at the catheter tip, when administered intrathecally. To further evaluate its potential neurotoxicity, we administered dynorphin A(1-13) to rats at very high doses IV. Rats (n = 6-10 per group) received dynorphin A(1-13) as bolus IV doses of 5 mg/kg, or as continuous IV infusions of 40 mg/kg/day for 1 day, with saline controls. The appearance and behavior of all animals was normal. Tail flick latencies remained unchanged (p > 0.5). There were no histologic abnormalities of the spinal cord or brain when examined by light microscopy. Two additional groups received bolus injections of dynorphin A(1-13) 50 or 100 mg/kg IV. Animals receiving 50 mg/kg showed cutaneous flushing, labored respirations, and decreased spontaneous movement, which resolved within 10 min. Histology at 1 week was normal. All six animals receiving 100 mg/kg convulsed and died within minutes. Three animals that received dynorphin A(1-13) 40 mg/kg/day for 7 days had normal behavior and histology. We conclude that the previously observed neurotoxicity of intrathecally administered dynorphin A(1-13) is a local effect that does not occur when dynorphin A(1-13) is administered IV, even at very high doses.


Asunto(s)
Analgésicos Opioides/toxicidad , Sistema Nervioso Central/patología , Dinorfinas/toxicidad , Dimensión del Dolor/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Animales , Conducta Animal/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Dinorfinas/administración & dosificación , Dinorfinas/farmacocinética , Infusiones Intravenosas , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacocinética , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos
13.
Eur J Pharmacol ; 261(3): 295-301, 1994 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-7813551

RESUMEN

Lumbar subarachnoid injection of dynorphin A causes an ischemia-induced neuronal degeneration and persistent hindlimb paralysis. The protective effects of a variety of competitive and non-competitive N-methyl-D-aspartate (NMDA) receptor antagonists indicate that activation of the NMDA receptor complex is essential for dynorphin A-induced spinal cord injury. 1-Aminocyclopropanecarboxylic acid (ACPC) is a high affinity, partial agonist at strychnine-insensitive glycine receptors associated with the NMDA receptor complex. Pretreatment of rats with ACPC (100 and 200 mg/kg, i.p., 30 min prior to dynorphin A) significantly eliminated the persistent hindlimb motor deficits and neuropathological changes produced by 20 nmol of this peptide. The neuroprotective effects of ACPC (100 mg/kg, i.p.) were abolished by parenteral administration of glycine (800 mg/kg, 30 min prior to ACPC), consistent with other in vivo and in vitro studies indicating that the pharmacological actions of ACPC are effected through strychnine-insensitive glycine receptors. When given instead as six daily injections (200 mg/kg, i.p.) followed by an injection-free day, ACPC also significantly improved neurological recovery following dynorphin-A injection. These results support earlier indications that: (1) activation of the NMDA receptor complex plays a critical role in mediating dynorphin A-induced rat spinal cord injury; (2) ACPC provides an effective means of antagonizing excitotoxic phenomena; and (3) chronic administration of ACPC can elicit a persistent change in the NMDA receptor complex.


Asunto(s)
Aminoácidos Cíclicos , Aminoácidos/farmacología , Dinorfinas/antagonistas & inhibidores , Enfermedades de la Médula Espinal/prevención & control , Animales , Dinorfinas/administración & dosificación , Dinorfinas/toxicidad , Miembro Posterior/fisiología , Inyecciones , Masculino , Parálisis/inducido químicamente , Parálisis/prevención & control , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Enfermedades de la Médula Espinal/inducido químicamente , Enfermedades de la Médula Espinal/patología , Espacio Subaracnoideo
14.
J Pharmacol Exp Ther ; 269(1): 358-66, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7909561

RESUMEN

Dynorphin A reduced lumbosacral blood flow, elevated cerebrospinal fluid lactic acid concentrations and caused flaccid hindlimb paralysis and striking neuropathological changes after its injection into the spinal subarachnoid space in rats. Coadministration of the vasodilator hydralazine substantially eliminated the paralytic, anaerobic metabolic and neuropathological responses to dynorphin A. In contrast, in concentrations up to 1 mM, dynorphin A did not alter the viability of cultured rat spinal cord neurons. Thus, it appears that this peptide lacks direct neurotoxic effects and that neuronal injuries in vivo result primarily from ischemia associated with dynorphin A-induced blood flow reductions. NMDA receptor antagonists significantly improved recovery from dynorphin A-induced hindlimb paralysis, and substantially eliminated neuropathological changes without attenuating the acute blood flow reductions or lactic acid elevations. Additionally, glutamate and aspartate concentrations were increased significantly in spinal cord cerebrospinal fluid samples removed during the time that peptide-induced spinal cord blood flow reductions were observed. In contrast, neither amino acid concentration was elevated in media removed after 1-hr exposure of spinal cord neuronal cell cultures to 100 microM concentrations of dynorphin A. These results indicate that the paralysis and spinal cord injuries produced in rats after spinal subarachnoid injection of dynorphin A result predominantly from spinal cord ischemia, and further identify excitatory amino acids and N-methyl-D-aspartate receptor mechanisms as important mediators in this injury model.


Asunto(s)
Aminoácidos/fisiología , Modelos Animales de Enfermedad , Dinorfinas/toxicidad , Isquemia/inducido químicamente , Receptores de N-Metil-D-Aspartato/fisiología , Traumatismos de la Médula Espinal/inducido químicamente , Médula Espinal/irrigación sanguínea , Animales , Células Cultivadas , Miembro Posterior/efectos de los fármacos , Miembro Posterior/fisiología , Masculino , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Médula Espinal/citología , Médula Espinal/efectos de los fármacos
15.
Pol J Pharmacol Pharm ; 44(2): 109-20, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1357636

RESUMEN

A novel analog of dynorphin (1-13), D-Ala2,F5Phe4-dynorphin amide, was prepared and its pharmacological spectrum of activity was investigated. In a hot plate test on Swiss Webster and C57Bl mice, a 20 micrograms intracerebroventricular (icv) dose of the analog produced analgesia, which was greater in potency and duration than the parent dynorphin. This action of D-Ala2,F5Phe4-dynorphin amide was antagonized by the opiate receptor antagonist naloxone (2 mg/kg ip), administered either before or after the peptide. In addition to its analgesic action in mice, D-Ala2,F5Phe4-dynorphin amide produced a Straub tail and a catatonic-like state, both of which were also attenuated by naloxone. On the electrically-stimulated mouse vas deferens preparation, in vitro, D-Ala2,F5Phe4-dynorphin amide inhibited contractile activity and had an IC50 of 108.2 +/- 34.7 nM (SEM), about 4-fold weaker than that of dynorphin. This action was also attenuated by naloxone. An icv dose of 150 micrograms of D-Ala2,F5Phe4-dynorphin amide in mice, and a cumulative series of icv doses up to 2600 micrograms in anesthetized rats, failed to produce a lethal effect. No pathological changes were observed in mouse liver and kidney at 24 h after a 50 mg/kg dose of the peptide analog. In rats anesthetized with diallylbarbital (70 mg/kg ip) and urethane (280 mg/kg ip), D-Ala2,F5Phe4-dynorphin amide did not modify blood pressure, heart rate and respiratory rate. However, when mice were injected peripherally with single doses of D-Ala2,F5Phe4-dynorphin amide, convulsive episodes were produced, and lethal effects were observed with an LD50 of 60.0 mg/kg (95% confidence limits: 49.7-70.2 mg/kg) at 48 h. This action of D-Ala2,F5Phe4-dynorphin amide was not attenuated by naloxone (2.0 mg/kg, ip). Although analgesic and behavioral effects of D-Ala2,F5Phe4-dynorphin amide (e.g. Straub tail and catatonic-like state) are opiate-like, the lethal effect may be the consequence of actions of the peptide on non-opiate systems, Thus, the novel fluorinated dynorphin analog, D-Ala2,F5Phe4-dynorphin amide, may be a useful chemical tool for the study of opiate systems and their occasionally unanticipated biological or toxic actions.


Asunto(s)
Analgésicos/toxicidad , Encéfalo/efectos de los fármacos , Dinorfinas/análogos & derivados , Fragmentos de Péptidos/toxicidad , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Dinorfinas/antagonistas & inhibidores , Dinorfinas/toxicidad , Femenino , Hemodinámica/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Naloxona/farmacología , Nociceptores/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Conducto Deferente/efectos de los fármacos
16.
Neurosci Lett ; 110(1-2): 113-7, 1990 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-1970139

RESUMEN

Intrathecal (i.t.) administration of the opioid dynorphin causes neurological dysfunction and tissue damage. It has been suggested that these effects of dynorphin may be mediated, in part, by N-methyl-D-aspartate (NMDA) receptors. In the present studies, recently developed compounds that block the glycine potentiation site of the NMDA receptor (Gly-NMDA site), including the competitive antagonist 5-fluoro-indole-2-carboxylic acid and the non-competitive antagonist 7-chlorokynurenic acid, prevented the neurologic deficits and mortality caused by i.t. dynorphin A(1-17). These findings are consistent with the hypothesis that dynorphin-induced neurological dysfunction involves activation of NMDA receptors. Moreover, blockade of the Gly-NMDA site may provide an alternative to blockade of the glutamate binding site or NMDA receptor ion channel as an in vivo pharmacological strategy to treat conditions previously associated with excitotoxin mediated tissue injury.


Asunto(s)
Dinorfinas/toxicidad , Glicina/farmacología , Indoles/farmacología , Ácido Quinurénico/análogos & derivados , Parálisis/inducido químicamente , Receptores de Neurotransmisores/fisiología , Animales , Indoles/uso terapéutico , Inyecciones Espinales , Ácido Quinurénico/farmacología , Ácido Quinurénico/uso terapéutico , Masculino , Parálisis/tratamiento farmacológico , Parálisis/metabolismo , Ratas , Ratas Endogámicas , Receptores de N-Metil-D-Aspartato , Receptores de Neurotransmisores/efectos de los fármacos , Receptores de Neurotransmisores/metabolismo
17.
Brain Res ; 497(1): 155-62, 1989 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-2571391

RESUMEN

The selective kappa opioid receptor antagonist nor-binaltorphimine (nor-BNI) was used to distinguish a kappa opioid component in the mechanisms underlying the hindlimb paralysis, ischemia, and neuronal injury induced in the rat by the kappa opioid agonist dynorphin A. Spinal intrathecal (i.t.) injection of nor-BNI (20 nmol) either 15 min or immediately before i.t. injections of 5 or 20 nmol of dynorphin A failed to alter the dynorphin A-induced disruption of hindlimb motor function and nociceptive responsiveness. Nor-BNI also did not change the 3-fold increases in cerebrospinal fluid lactate concentrations produced by 20 nmol of dynorphin A. Neuroanatomical evaluations revealed that the cell loss, fiber degeneration, and central gray necrosis in lumbosacral spinal cords of rats treated with 20 nmol of dynorphin A were not altered by nor-BNI (20 nmol, i.t.). Thus, the spinal cord injury and associated neurological deficits resulting from i.t. injection of dynorphin A appear to be primarily, if not totally, attributable to its non-kappa opioid action(s).


Asunto(s)
Dinorfinas/toxicidad , Isquemia/inducido químicamente , Naltrexona/análogos & derivados , Parálisis/inducido químicamente , Receptores Opioides/fisiología , Enfermedades de la Médula Espinal/inducido químicamente , Animales , Atrofia , Miembro Posterior , Isquemia/metabolismo , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos , Parálisis/metabolismo , Parálisis/patología , Ratas , Ratas Endogámicas , Receptores Opioides kappa , Enfermedades de la Médula Espinal/metabolismo , Enfermedades de la Médula Espinal/patología
18.
Life Sci ; 44(20): 1505-14, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2566882

RESUMEN

Intrathecally injected dynorphin A (1-13) in rats results in a reversible hindlimb paralysis and an irreversible loss of the tail-flick reflex. Histologic examination of the spinal cords of dynorphin treated rats demonstrated dead and/or dying neurons predominately localized in the central area which approximates Rexed lamina VII and X. In this area a maximum effect of the dynorphin-induced neurotoxicity is evident. Thus, the dynorphin-induced neuron death is suggestive of an anatomical selectivity.


Asunto(s)
Dinorfinas/toxicidad , Fragmentos de Péptidos/toxicidad , Enfermedades de la Médula Espinal/inducido químicamente , Animales , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Dinorfinas/administración & dosificación , Masculino , Neuronas/patología , Dolor , Parálisis/inducido químicamente , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Endogámicas , Reflejo , Enfermedades de la Médula Espinal/patología
19.
Eur J Pharmacol ; 153(1): 45-54, 1988 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-2905665

RESUMEN

Dynorphin A-(1-17) acts through non-opioid mechanisms to produce dose-related neurological deficits following injection into the lumbar spinal subarachnoid space in rats. Hindlimb motor function was examined following subarachnoid injection of dynorphin A fragments and other opioid peptides derived from prodynorphin to establish: (1) which portion(s) of the dynorphin A molecule cause hindlimb motor dysfunction, and (2) whether these paralytic actions are shared by other opioids (dynorphin B, alpha-neo-endorphin, and beta-neo-endorphin) derived from the same promolecule. To minimize the influence of enzymatic inactivation on relative bioactivities, peptides were coinjected with a combination of peptidase inhibitors previously shown to enhance the actions of dynorphin A fragments in vitro. Dynorphin A-(1-17) and -(2-17) produced dose-related neurological deficits with equal potencies and durations. Although without effect when injected alone, dynorphin A-(1-8), -(1-7) and -(3-8) caused transient motor dysfunction when co-injected with peptidase inhibitors. In contrast, dynorphin A-(1-6), -(1-5) and -(6-17) did not disrupt hindlimb motor function with or without peptidase inhibition. Dynorphin B, alpha-neo-endorphin and beta-neo-endorphin also caused hindlimb dysfunction which was potentiated by peptidase inhibition. These deficits appeared to result from non-opioid actions of these three peptides, since they were not blocked by the opioid antagonist naloxone. Thus, the paralytic effects of dynorphin A: (1) result from non-opioid actions involving the 3-7 or 3-8 positions of the molecule, and (2) are shared by other prodynorphin-derived opioid peptides.


Asunto(s)
Dinorfinas/toxicidad , Parálisis/inducido químicamente , Animales , Dinorfinas/administración & dosificación , Miembro Posterior/efectos de los fármacos , Inyecciones , Masculino , Naloxona/farmacología , Ratas , Ratas Endogámicas , Médula Espinal , Espacio Subaracnoideo
20.
J Pharmacol Exp Ther ; 244(3): 1169-77, 1988 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2908046

RESUMEN

The delta opioid receptor antagonist ICI 174864 produces postural abnormalities and barrel rolling after i.c.v. injection and hindlimb and tail flaccidity after spinal subarachnoid injection in rats. These effects appear to result from nonopioid characteristics of ICI 174864 because they are neither shared nor blocked by other opioid antagonists (naloxone, ICI 154129 and WIN 44,441-3) and are produced by two compounds (ICI 174644 and ICI 178173) that are structurally related to ICI 174864 but lack its delta antagonist properties. Barrel rolling and hindlimb paralysis are also produced by dynorphin A-related peptides; however, rats failed to demonstrate tolerance or cross-tolerance to the hindlimb paralytic actions of ICI 174864 or dynorphin A (1-13) after 7 days of continuous spinal intrathecal infusion of either of these compounds. Whereas hindlimb responses to low doses of ICI 174864 (1.6-6.2 nmol intrathecally) were usually transient, higher doses (6.2-25 nmol intrathecally) produced persistent hindlimb motor dysfunction, altered nociception, priapism, hindlimb edema, bladder infarction and atony and urinary incontinence. Neuronal and axonal changes in the lumbosacral spinal cords of rats with persistent and transient neurologic deficits provided direct evidence of the neuropathologic actions of ICI 174864 (3.1 and 6.2 nmol) and ICI 174644 (25 nmol). These results indicate that 1) use of ICI 174864 as a selective delta opioid receptor antagonist is potentially compromised by its nonopioid neuropathologic actions and 2) ICI 174864 and dynorphin A-related peptides are unique among opioid agonists and antagonists in sharing barrel rolling and hindlimb paralytic effects. A similar mechanism of action may underlie the shared nonopioid actions of these peptides.


Asunto(s)
Encefalina Leucina/análogos & derivados , Antagonistas de Narcóticos/toxicidad , Neuronas/efectos de los fármacos , Parálisis/inducido químicamente , Receptores Opioides/efectos de los fármacos , Animales , Tolerancia a Medicamentos , Dinorfinas/toxicidad , Encefalina Leucina/toxicidad , Masculino , Ratas , Ratas Endogámicas , Receptores Opioides delta , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA