Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Res ; 1739: 146858, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32348776

RESUMEN

PACAP-38 (P38) is a pleiotropic peptide that exerts multiple peripheral and central actions, including neurotrophic, neuroprotective and anti-inflammatory actions. Previous studies have suggested an improvement of memory in rats that have received a single systemic injection of P38. In a therapeutic perspective, we used an analog, acetyl-[Ala15, Ala20]PACAP-38-propylamide (ALG), to improve both stability and affinity for PAC1 receptors vs. endogen PACAP. We investigated the effect of P38 and ALG on memory consolidation using a spatial novelty detection (SND) task in which rats had to memorize a configuration of objects to identify that, during a test session, a familiar object has been moved to a new location. Rats received an intravenous injection of P38 or ALG after the last training session. In Experiment 1, P38 (30 µg/kg) improved spatial memory consolidation allowing detection of novelty vs. saline injection. In Experiment 2, we confirmed this effect and showed that P38 restored the performance similar to what was found using non-injected rats. This suggests that, contrary to ALG, P38 exerted a promesiant rather than an anxiety-related effect whereas ALG did not show similar action. We also examined whether P38 effect involved an interaction with NR2B-containing NMDA receptors (NMDARs) by administrating ifenprodil (IFE; a selective NR2B-containing NMDAR antagonist) alone or in combination with P38 or ALG. The results suggested that P38 action on memory involved NR2B-containing NMDARs. Lastly, brain-derived neutrophic factor (BDNF) modulation appeared to be not related to the behavioral performance in the SND task. Overall, the results indicate that P38 exerted a beneficial effect on memory consolidation in a non-associative task, whereas ALG did not have this action.


Asunto(s)
Consolidación de la Memoria/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Memoria Espacial/efectos de los fármacos , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Consolidación de la Memoria/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Ratas , Ratas Wistar , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo
2.
Biochem Biophys Res Commun ; 503(3): 1973-1979, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30077368

RESUMEN

Vasoactive intestinal peptide receptor 2 (VIPR2, also known as VPAC2) is a class B G-protein coupled receptor (GPCR) and plays important roles in the physiology of central nervous system (CNS) by interaction with natural ligands; vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP). Because it has been reported that high-expression and/or overactivation of VIPR2 link to schizophrenic symptoms, VIPR2 antagonists could be good drug candidates for schizophrenia therapeutics. In this study, we discovered several artificial peptides that antagonize both human and rodent VIPR2 with selectivities against receptor subtypes VIPR1 (also known as VPAC1) and pituitary adenylate cyclase-activating polypeptide type-1 receptor (PAC1). Of them, the representative 16-mer cyclic peptide VIpep-3 (Ac-CPPYLPRRLCTLLLRS-OH) exhibited strong binding affinity with KD value of 41 nM to extracellular domain of human VIPR2 in SPR analysis and showed potent antagonist activity with IC50 values of 47 nM (human), 180 nM (mouse), and 44 nM (rat) against VIP-VIPR2 signal in cell-based Ca influx assay. This is not only the first report on artificial VIPR2-selective antagonist peptides but also good example of the effective approach to discover novel antagonist against class B GPCR. Our peptides will contribute to study and development of the novel CNS drugs targeting to VIPR2.


Asunto(s)
Descubrimiento de Drogas , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Péptido Intestinal Vasoactivo/farmacología , Animales , Técnicas Biosensibles , Células CHO , Cricetulus , Humanos , Ligandos , Ratones , Biblioteca de Péptidos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Ratas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Péptido Intestinal Vasoactivo/síntesis química , Péptido Intestinal Vasoactivo/química
3.
Peptides ; 78: 30-41, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26826611

RESUMEN

Neuroblastoma (NB) is a pediatric cancer. New therapies for high-risk NB aim to induce cell differentiation and to inhibit MYCN and ALK signaling in NB. The vasoactive intestinal peptide (VIP) and the pituitary adenylate cyclase-activating polypeptide (PACAP) are 2 related neuropeptides sharing common receptors. The level of VIP increases with NB differentiation. Here, the effects of VIP and PACAP analogs developed for therapeutic use were studied in MYCN-amplified NB SK-N-DZ and IMR-32 cells and in Kelly cells that in addition present the F1174L ALK mutation. As previously reported by our group in IMR-32 cells, VIP induced neuritogenesis in SK-N-DZ and Kelly cells and reduced MYCN expression in Kelly but not in SK-N-DZ cells. VIP decreased AKT activity in the ALK-mutated Kelly cells. These effects were PKA-dependent. IMR-32, SK-NDZ and Kelly cells expressed the genes encoding the 3 subtypes of VIP and PACAP receptors, VPAC1, VPAC2 and PAC1. In parallel to its effect on MYCN expression, VIP inhibited invasion in IMR-32 and Kelly cells. Among the 3 PACAP analogs tested, [Hyp(2)]PACAP-27 showed higher efficiency than VIP in Kelly cells. These results indicate that VIP and PACAP analogs act on molecular and cellular processes that could reduce aggressiveness of high-risk NB.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Péptido Intestinal Vasoactivo/farmacología , Quinasa de Linfoma Anaplásico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Mutación , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuronas/metabolismo , Neuronas/patología , Especificidad de Órganos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Péptido Intestinal Vasoactivo/síntesis química
4.
PLoS One ; 9(3): e91541, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24643018

RESUMEN

Pituitary adenylate cyclase activating polypeptide (PACAP) is an important neurotrophic factor influencing differentiation of neuronal elements and exerting protecting role during traumatic injuries or inflammatory processes of the central nervous system. Although increasing evidence is available on its presence and protecting function in various peripheral tissues, little is known about the role of PACAP in formation of skeletal components. To this end, we aimed to map elements of PACAP signalling in developing cartilage under physiological conditions and during oxidative stress. mRNAs of PACAP and its receptors (PAC1,VPAC1, VPAC2) were detectable during differentiation of chicken limb bud-derived chondrogenic cells in micromass cell cultures. Expression of PAC1 protein showed a peak on days of final commitment of chondrogenic cells. Administration of either the PAC1 receptor agonist PACAP 1-38, or PACAP 6-38 that is generally used as a PAC1 antagonist, augmented cartilage formation, stimulated cell proliferation and enhanced PAC1 and Sox9 protein expression. Both variants of PACAP elevated the protein expression and activity of the Ca-calmodulin dependent Ser/Thr protein phosphatase calcineurin. Application of PACAPs failed to rescue cartilage formation when the activity of calcineurin was pharmacologically inhibited with cyclosporine A. Moreover, exogenous PACAPs prevented diminishing of cartilage formation and decrease of calcineurin activity during oxidative stress. As an unexpected phenomenon, PACAP 6-38 elicited similar effects to those of PACAP 1-38, although to a different extent. On the basis of the above results, we propose calcineurin as a downstream target of PACAP signalling in differentiating chondrocytes either in normal or pathophysiological conditions. Our observations imply the therapeutical perspective that PACAP can be applied as a natural agent that may have protecting effect during joint inflammation and/or may promote cartilage regeneration during degenerative diseases of articular cartilage.


Asunto(s)
Calcineurina/genética , Condrocitos/efectos de los fármacos , Condrogénesis/genética , Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , Calcineurina/metabolismo , Inhibidores de la Calcineurina/farmacología , Diferenciación Celular , Proliferación Celular , Embrión de Pollo , Condrocitos/citología , Condrocitos/metabolismo , Ciclosporina/farmacología , Regulación del Desarrollo de la Expresión Génica , Peróxido de Hidrógeno/farmacología , Esbozos de los Miembros/citología , Esbozos de los Miembros/efectos de los fármacos , Esbozos de los Miembros/crecimiento & desarrollo , Esbozos de los Miembros/metabolismo , Estrés Oxidativo , Péptidos/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Cultivo Primario de Células , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal
5.
Peptides ; 32(6): 1207-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21514338

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) shows potent protective effects in numerous models of neurological insults. However, the use of PACAP as a clinically efficient drug is limited by its poor metabolic stability. By combining identification of enzymatic cleavage sites with targeted chemical modifications, a metabolically stable and potent PACAP38 analog was recently developed. The neuroprotective activity of this novel compound was for the first time evaluated and compared to the native peptide using a rat model of middle cerebral artery occlusion (MCAO). Our results show that as low as picomolar doses of PACAP38 and its analog strongly reduce infarct volume and improve neurological impairment induced by stroke. In particular, these peptides inhibit the expression of Bcl-2-associated death promoter, caspase 3, macrophage inflammatory protein-1α, inducible nitric oxide synthase 2, tumor necrosis factor-α mRNAs, and increase extracellular signal-regulated kinase 2, B-cell CLL/lymphoma 2 and interleukin 6 mRNA levels. These results indicate that the neuroprotective effect of PACAP after MCAO is not only due to its ability to inhibit apoptosis but also to modulate the inflammatory response. The present study highlights the potential therapeutic efficacy of very low concentrations of PACAP or its metabolically stable derivative for the treatment of stroke.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia/patología , Fármacos Neuroprotectores/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Accidente Cerebrovascular/patología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Expresión Génica , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Proteínas Inflamatorias de Macrófagos/genética , Proteínas Inflamatorias de Macrófagos/metabolismo , Masculino , Fármacos Neuroprotectores/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , ARN Mensajero/análisis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
6.
J Mol Neurosci ; 43(1): 85-93, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20585898

RESUMEN

Pituitary adenylate cyclase activating polypeptide 38 (PACAP38), one of the major peptide transmitters, has emerged as a promising drug candidate for the treatment of type 2 diabetes. In the present study, on the basis of previous structure-activity relationships, a new PACAP38 derivative, [R(15, 20, 21), L(17)]-PACAP38, was chemically synthesized with the aim of enhancing the therapeutic potential of PACAP38. The solution structure of the new derivative was almost identical to that of PACAP38 as evaluated by circular dichroic spectroscopy, and both PACAP38 and the new derivative stimulated adenylate cyclase in rat insulinoma RIN-m5F cells with EC(50) values of 4.6 and 5.5 nM, respectively. Stability studies revealed the gradual degradation of PACAPs in rat serum, although there appeared to be a 42% reduction in degradation kinetics for [R(15, 20, 21), L(17)]-PACAP38 compared with that of PACAP38. The novel derivative also exhibited more potent protective effects against streptozotocin (STZ)-induced apoptotic death of RIN-m5F cells, possibly due to the enhanced stability. The n0-STZ model, in which neonatal rats were injected with STZ at birth, developed a typical diabetic condition; however, chronic administration of [R(15, 20, 21), L(17)]-PACAP38 resulted in protection of pancreatic islets, followed by the improvement of glycemic control. Thus, the chemical modification of PACAP38 led to the development of a new promising derivative with enhanced stability and biological activity, and early administration of [R(15, 20, 21), L(17)]-PACAP38 might be of help for preventing the development of diabetes in type 2 diabetic model rats.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Estabilidad Proteica , Animales , Dicroismo Circular , Diabetes Mellitus Experimental/tratamiento farmacológico , Prueba de Tolerancia a la Glucosa , Masculino , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Ratas , Ratas Wistar
7.
Peptides ; 29(6): 919-32, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18353507

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38- or 27-amino acid neuropeptide with promising therapeutic applications for the treatment of several pathophysiological states related to neurodegenerative diseases. However, its use for therapeutic applications is actually limited by its restricted bioavailability and rapid degradation. Therefore, metabolically stable PACAP analogs represent promising tools to further investigate the physiological roles of PACAP and ascertain its usefulness in some clinical conditions. In this study, derivatives of PACAP27 and PACAP38 have been rationally designed to develop PAC1 receptor agonists resistant to peptidase action. Results showed that N-terminal modifications confer resistance to dipeptidyl peptidase IV, a major proteolytic process involved in PACAP degradation. Moreover, in vitro incubation of both PACAP isoforms in human plasma revealed that PACAP38 is rapidly metabolized, with a half-life of less than 5 min, while PACAP27 was stable in these experimental conditions. Hence, following the elucidation of its plasmatic metabolites, PACAP38 was modified at its putative endopeptidase and carboxypeptidase sites of cleavage. All peptide analogs were tested for their ability to bind the PAC1 receptor, as well as for their potency to induce calcium mobilization and inhibit PC12 cell proliferation through the PAC1 receptor. This approach revealed two leading compounds, i.e. acetyl-[Ala15, Ala20]PACAP38-propylamide and acetyl-PACAP27-propylamide, which exhibited improved metabolic stability and potent biological activity. This study describes innovative data related to PACAP metabolism in human plasma and depicts the development of a metabolically stable PACAP38 analog, acetyl-[Ala15, Ala20]PACAP38-propylamide, which behaves as a super-agonist towards the PAC1 receptor.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células CHO , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Cricetinae , Cricetulus , ADN Complementario , Relación Dosis-Respuesta a Droga , Electroporación , Humanos , Datos de Secuencia Molecular , Células PC12 , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/sangre , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/aislamiento & purificación , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Factores de Tiempo
8.
Ann N Y Acad Sci ; 1070: 365-70, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16888193

RESUMEN

The present article investigated effects of systemic pituitary adenylate cyclase-activating polypeptide (PACAP) treatment in monosodium glutamate (MSG)-induced retinal degeneration and neurobehavioral alterations in neonatal rats. It was found that the dose of PACAP that effectively enhances neurobehavioral development in normal rats was able to counteract the retarding effect of MSG on righting, forelimb placing, and grasp reflexes and caused a significant amelioration of the righting and gait reflex performance and motor coordination at 2 weeks of age. In the retina, significant amelioration of neuronal loss in the inner retinal layers was achieved, but it was much less than that observed by local administration.


Asunto(s)
Conducta Animal/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/tratamiento farmacológico , Glutamato de Sodio/farmacología , Animales , Peso Corporal/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/síntesis química , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...