Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Front Endocrinol (Lausanne) ; 12: 732456, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759890

RESUMEN

Mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP) display psychomotor abnormalities, most of which are ameliorated by atypical antipsychotics with serotonin (5-HT) 2A receptor (5-HT2A) antagonism. Heterozygous Pacap mutant mice show a significantly higher hallucinogenic response than wild-type mice to a 5-HT2A agonist. Endogenous PACAP may, therefore, affect 5-HT2A signaling; however, the underlying neurobiological mechanism for this remains unclear. Here, we examined whether PACAP modulates 5-HT2A signaling by addressing cellular protein localization. PACAP induced an increase in internalization of 5-HT2A but not 5-HT1A, 5-HT2C, dopamine D2 receptors or metabotropic glutamate receptor 2 in HEK293T cells. This PACAP action was inhibited by protein kinase C inhibitors, ß-arrestin2 silencing, the PACAP receptor PAC1 antagonist PACAP6-38, and PAC1 silencing. In addition, the levels of endogenous 5-HT2A were decreased on the cell surface of primary cultured cortical neurons after PACAP stimulation and were increased in frontal cortex cell membranes of Pacap-/- mice. Finally, intracerebroventricular PACAP administration suppressed 5-HT2A agonist-induced head twitch responses in mice. These results suggest that PACAP-PAC1 signaling increases 5-HT2A internalization resulting in attenuation of 5-HT2A-mediated signaling, although further study is necessary to determine the relationship between behavioral abnormalities in Pacap-/- mice and PACAP-induced 5-HT2A internalization.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptor de Serotonina 5-HT2A/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Transporte de Proteínas/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Transducción de Señal/fisiología
2.
Neuroendocrinology ; 111(1-2): 45-69, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32028278

RESUMEN

OBJECTIVE: We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS: Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS: In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS: These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Metabolismo Energético/fisiología , Neuronas/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Proopiomelanocortina , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Fenómenos Electrofisiológicos , Metabolismo Energético/efectos de los fármacos , Femenino , Cobayas , Homeostasis , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/efectos de los fármacos
3.
J Headache Pain ; 19(1): 28, 2018 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-29619773

RESUMEN

Pituitary adenylate-cyclase-activating polypeptide (PACAP) is a 27- or 38-amino acid neuropeptide, which belongs to the vasoactive intestinal polypeptide (VIP)/glucagon/secretin family. PACAP shows particularly high homology (~ 68%) to VIP. Because of the high homology of the amino acid sequences of PACAP and VIP, these peptides share three class B-G-protein coupled receptors: the PAC1-Receptor (PAC1-R), the VPAC1-Receptor (VPAC1-R) and VPAC2-Receptor (VPAC2-R). These receptors have high homology to each other, and their high homology is utilized for these discoveries. This review provides mainly an overview of the history of the discovery of PACAP and its three receptors.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Animales , Humanos
4.
Br J Dermatol ; 176(2): 413-422, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27453364

RESUMEN

BACKGROUND: Sweat secretion is the major function of eccrine sweat glands; when this process is disturbed (paridrosis), serious skin problems can arise. To elucidate the causes of paridrosis, an improved understanding of the regulation, mechanisms and factors underlying sweat production is required. Pituitary adenylate cyclase-activating polypeptide (PACAP) exhibits pleiotropic functions that are mediated via its receptors [PACAP-specific receptor (PAC1R), vasoactive intestinal peptide (VIP) receptor type 1 (VPAC1R) and VPAC2R]. Although some studies have suggested a role for PACAP in the skin and several exocrine glands, the effects of PACAP on the process of eccrine sweat secretion have not been examined. OBJECTIVES: To investigate the effect of PACAP on eccrine sweat secretion. METHODS: Reverse transcriptase-polymerase chain reaction and immunostaining were used to determine the expression and localization of PACAP and its receptors in mouse and human eccrine sweat glands. We injected PACAP subcutaneously into the footpads of mice and used the starch-iodine test to visualize sweat-secreting glands. RESULTS: Immunostaining showed PACAP and PAC1R expression by secretory cells from mouse and human sweat glands. PACAP immunoreactivity was also localized in nerve fibres around eccrine sweat glands. PACAP significantly promoted sweat secretion at the injection site, and this could be blocked by the PAC1R-antagonist PACAP6-38. VIP, an agonist of VPAC1R and VPAC2R, failed to induce sweat secretion. CONCLUSIONS: This is the first report demonstrating that PACAP may play a crucial role in sweat secretion via its action on PAC1R located in eccrine sweat glands. The mechanisms underlying the role of PACAP in sweat secretion may provide new therapeutic options to combat sweating disorders.


Asunto(s)
Glándulas Ecrinas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Sudor/metabolismo , Adulto , Animales , Femenino , Pie , Humanos , Masculino , Ratones Endogámicos C57BL , Fibras Nerviosas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , ARN Mensajero/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/fisiología , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/fisiología
5.
J Pharmacol Sci ; 130(4): 194-203, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26948958

RESUMEN

Intrathecal (i.t.) administration of pituitary adenylate cyclase-activating polypeptide (PACAP) induces long-lasting nociceptive behaviors for more than 60 min in mice, while the involvement of PACAP type1 receptor (PAC1-R) has not been clarified yet. The present study investigated signaling mechanisms of the PACAP-induced prolonged nociceptive behaviors. Single i.t. injection of a selective PAC1-R agonist, maxadilan (Max), mimicked nociceptive behaviors in a dose-dependent manner similar to PACAP. Pre- or post-treatment of a selective PAC1-R antagonist, max.d.4, significantly inhibited the nociceptive behaviors by PACAP or Max. Coadministration of a protein kinase A inhibitor, Rp-8-Br-cAMPS, a mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase inhibitor, PD98059 or a c-Jun N-terminal kinase (JNK) inhibitor, SP600125, significantly inhibited the nociceptive behaviors by Max. Immunohistochemistry and immunoblotting analysis revealed that spinal administration of Max-induced ERK phosphorylation and JNK phosphorylation, and also augmented an astrocyte marker, glial fibrillary acidic protein in mouse spinal cord. Furthermore, an astroglial toxin, l-α-aminoadipate, significantly attenuated the development of the nociceptive behaviors and ERK phosphorylation by Max. These results suggest that the activation of spinal PAC1-R induces long-lasting nociception through the interaction of neurons and astrocytes.


Asunto(s)
Astrocitos/fisiología , Conducta Animal/fisiología , Nocicepción/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Transducción de Señal , Médula Espinal/citología , Médula Espinal/fisiología , Animales , Masculino , Ratones Endogámicos
6.
Peptides ; 79: 39-48, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26976270

RESUMEN

PACAP-27 and PACAP-38 are the exclusive physiological ligands for the mammalian PAC1 receptor. The role of C-terminal amidation of these ligands at that receptor was examined in neuroendocrine cells expressing the PAC1 receptor endogenously and in non-neuroendocrine cells in which the human and rat PAC1 receptors were expressed from stable single-copy genes driven by the CMV promoter, providing stoichiometrically appropriate levels of this Gs-coupled GPCR in order to examine the potency and intrinsic activity of PACAP ligands and their des-amidated congeners. We found that replacement of the C-terminal glycine residues of PACAP-27 and -38 with a free acid; or extension of either peptide with the two to three amino acids normally found at these positions in PACAP processing intermediates in vivo following endoproteolytic cleavage and after exoproteolytic trimming and glycine-directed amidated, were equivalent in potency to the fully processed peptides in a variety of cell-based assays. These included real-time monitoring of cyclic AMP generation in both NS-1 neuroendocrine cells and non-neuroendocrine HEK293 cells; PKA-dependent gene activation in HEK293 cells; and neuritogenesis and cell growth arrest in NS-1 cells. The specific implications for the role of amidation in arming of secretin-related neuropeptides for biological function, and the general implications for neuropeptide-based delivery in the context of gene therapy, are discussed.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Amidas/farmacología , Secuencia de Aminoácidos , Animales , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Ratas , Sistemas de Mensajero Secundario
7.
PLoS One ; 11(1): e0146981, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26757053

RESUMEN

Light signals from intrinsically photosensitive retinal ganglion cells (ipRGCs) entrain the circadian clock and regulate negative masking. Two neurotransmitters, glutamate and Pituitary Adenylate Cyclase Activating Polypeptide (PACAP), found in the ipRGCs transmit light signals to the brain via glutamate receptors and the specific PACAP type 1 (PAC1) receptor. Light entrainment occurs during the twilight zones and has little effect on clock phase during daytime. When nocturnal animals have access to food only for a few hours during the resting phase at daytime, they adapt behavior to the restricted feeding (RF) paradigm and show food anticipatory activity (FAA). A recent study in mice and rats demonstrating that light regulates FAA prompted us to investigate the role of PACAP/PAC1 signaling in the light mediated regulation of FAA. PAC1 receptor knock out (PAC1-/-) and wild type (PAC1+/+) mice placed in running wheels were examined in a full photoperiod (FPP) of 12:12 h light/dark (LD) and a skeleton photoperiod (SPP) 1:11:1:11 h L:DD:L:DD at 300 and 10 lux light intensity. Both PAC1-/- mice and PAC1+/+ littermates entrained to FPP and SPP at both light intensities. However, when placed in RF with access to food for 4-5 h during the subjective day, a significant change in behavior was observed in PAC1-/- mice compared to PAC1+/+ mice. While PAC1-/- mice showed similar FAA as PAC1+/+ animals in FPP at 300 lux, PAC1-/- mice demonstrated an advanced onset of FAA with a nearly 3-fold increase in amplitude compared to PAC1+/+ mice when placed in SPP at 300 lux. The same pattern of FAA was observed at 10 lux during both FPP and SPP. The present study indicates a role of PACAP/PAC1 signaling during light regulated FAA. Most likely, PACAP found in ipRGCs mediating non-image forming light information to the brain is involved.


Asunto(s)
Ritmo Circadiano , Conducta Alimentaria , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Animales , Conducta Animal , Encéfalo/fisiología , Femenino , Ácido Glutámico/química , Luz , Masculino , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Fotoperiodo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Células Ganglionares de la Retina/citología , Transducción de Señal , Núcleo Supraquiasmático/metabolismo
8.
Dialogues Clin Neurosci ; 18(4): 403-413, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28179812

RESUMEN

Trauma-related disorders, such as posttraumatic stress disorder (PTSD) are remarkably common and debilitating, and are often characterized by dysregulated threat responses. Across numerous epidemiological studies, females have been found to have an approximately twofold increased risk for PTSD and other stress-related disorders. Understanding the biological mechanisms of this differential risk is of critical importance. Recent data suggest that the pituitary adenylate cyclase-activating polypeptide (PACAP) pathway is a critical regulator of the stress response across species. Moreover, increasing evidence suggests that this pathway is regulated by both stress and estrogen modulation and may provide an important window into understanding mechanisms of sex differences in the stress response. We have recently shown that PACAP and its receptor (PAC1R) are critical mediators of abnormal processes after psychological trauma. Notably, in heavily traumatized human subjects, there appears to be a robust sex-specific association of PACAP blood levels and PAC1R gene variants with fear physiology, PTSD diagnosis, and symptoms, specifically in females. The sex-specific association occurs within a single-nucleotide polymorphism (rs2267735) that resides in a putative estrogen response element involved in PAC1R gene regulation. Complementing these human data, the PAC1R messenger RNA is induced with fear conditioning or estrogen replacement in rodent models. These data suggest that perturbations in the PACAP-PAC1R pathway are regulated by estrogen and are involved in abnormal fear responses underlying PTSD.


Los trastornos relacionados con el trauma, como el trastorno por estrés postraumático (TEPT) en humanos, son extraordinariamente comunes y desgastadores, y a menudo están caracterizados por respuestas desreguladas a la amenaza. En numerosos estudios epidemiológicos se ha encontrado que las mujeres tienen un riesgo aumentado al doble para el TEPT y otros trastornos relacionados con el estrés. La comprensión de los mecanismos biológicos de este riesgo diferencial es de gran importancia. Hay datos recientes que sugieren que, a través de las especies, la vía del polipéptido activador de la adenilato ciclasa hipofisiaria (PAACH) tiene una regulación central en la respuesta de estrés. Sin embargo, hay evidencia creciente que sugiere que esta vía está regulada por el estrés y la modulación estrogénica y puede aportar una ventana importante para la comprensión de los mecanismos de las diferencias por sexo en la respuesta de estrés. Nosotros hemos mostrado recientemente que el PAACH y su receptor (R1PAC) son importantes mediadores de los procesos anormales después del trauma psicológico. Es notable que, específicamente en mujeres víctimas de grandes traumas, al parecer hay una potente asociación específica entre los niveles sanguíneos de PAACH y variantes del gen R1PAC con la fisiología del miedo, y síntomas y diagnóstico de TEPT. La asociación específica con el sexo ocurre con el polimorfismo del nucleótido único (rs2267735) que se encuentra en un elemento de la respuesta putativa de estrógeno involucrada en la regulación del gen R1PAC. Como complemento a estos datos humanos, en modelos de roedores el ARN mensajero del R1PAC está inducido por el condicionamiento al miedo o por el reemplazo de estrógenos. Estos datos sugieren que las perturbaciones en la vía del PACCH-R1PAC están reguladas por estrógenos y participan en las respuestas anormales al miedo del TEPT.


Les troubles liés aux traumatismes, comme les troubles du stress post-traumatique (TSPT) chez les humains, sont extrêmement courants et invalidants et souvent caractérisés par une dérégulation des réponses à la menace. De nombreuses études épidémiologiques indiquent que les femmes ont environ deux fois plus de risque de TSPT et d'autres troubles liés au stress. La compréhension des mécanismes biologiques de ce risque différentiel est d'une importance essentielle. Selon des données récentes, la voie du PACAP (pituitary adenylate cyclase-activating polypeptide) est un régulateur capital de la réponse au stress commun à l'ensemble des espèces. De plus, il existe des preuves croissantes de la régulation de ces voies par la modulation à la fois du stress et des estrogènes et de leur apport d'une ouverture importante dans la compréhension du mécanisme des différences selon le sexe dans la réponse au stress. Nous avons récemment montré que la PACAP et son récepteur (PAC1R) sont des médiateurs essentiels des processus anormaux après un traumatisme psychologique. Il semble y avoir, en particulier chez les humains fortement traumatisés, une importante association des concentrations sanguines de PACAP et des variants du gène PAS1R spécifiques du sexe avec la physiologie de la peur, le diagnostic de TSPT et des symptômes, en particulier chez les femmes. L'association spécifique au sexe est liée à un polymorphisme nucléotidique (rs2267735) localisé dans une séquence d'ADN supposée être un élément de réponse à l'estrogène impliqué dans la régulation du gène PAC1R. Pour compléter ces données humaines, l'ARN messager du PAC1R est induit avec le conditionnement à la peur ou le remplacement des estrogènes dans des modèles murins. Ces données suggèrent que les perturbations dans la voie du PACAP-PAC1R sont régulées par les estrogènes et impliquées dans les réponses anormales à la peur sous-tendant le TSPT.


Asunto(s)
Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/fisiopatología , Adaptación Psicológica/fisiología , Animales , Biomarcadores/sangre , Estrógenos/fisiología , Miedo/fisiología , Femenino , Interacción Gen-Ambiente , Humanos , Masculino , Memoria/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/sangre , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polimorfismo Genético , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Factores de Riesgo , Caracteres Sexuales , Factores Sexuales , Trastornos por Estrés Postraumático/sangre , Trastornos por Estrés Postraumático/genética , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Estrés Psicológico/genética
9.
J Neurosci ; 35(39): 13430-47, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26424889

RESUMEN

Imprinted genes are dosage sensitive, and their dysregulated expression is linked to disorders of growth and proliferation, including fetal and postnatal growth restriction. Common sequelae of growth disorders include neurodevelopmental defects, some of which are indirectly related to placental insufficiency. However, several growth-associated imprinted genes are also expressed in the embryonic CNS, in which their aberrant expression may more directly affect neurodevelopment. To test whether growth-associated genes influence neural lineage progression, we focused on the maternally imprinted gene Zac1. In humans, either loss or gain of ZAC1 expression is associated with reduced growth rates and intellectual disability. To test whether increased Zac1 expression directly perturbs neurodevelopment, we misexpressed Zac1 in murine neocortical progenitors. The effects were striking: Zac1 delayed the transition of apical radial glial cells to basal intermediate neuronal progenitors and postponed their subsequent differentiation into neurons. Zac1 misexpression also blocked neuronal migration, with Zac1-overexpressing neurons pausing more frequently and forming fewer neurite branches during the period when locomoting neurons undergo dynamic morphological transitions. Similar, albeit less striking, neuronal migration and morphological defects were observed on Zac1 knockdown, indicating that Zac1 levels must be regulated precisely. Finally, Zac1 controlled neuronal migration by regulating Pac1 transcription, a receptor for the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP). Pac1 and Zac1 loss- and gain-of-function presented as phenocopies, and overexpression of Pac1 rescued the Zac1 knockdown neuronal migration phenotype. Thus, dysregulated Zac1 expression has striking consequences on neocortical development, suggesting that misexpression of this transcription factor in the brain in certain growth disorders may contribute to neurocognitive deficits. Significance statement: Altered expression of imprinted genes is linked to cognitive dysfunction and neuropsychological disorders, such as Angelman and Prader-Willi syndromes, and autism spectrum disorder. Mouse models have also revealed the importance of imprinting for brain development, with chimeras generated with parthenogenetic (two maternal chromosomes) or androgenetic (two paternal chromosomes) cells displaying altered brain sizes and cellular defects. Despite these striking phenotypes, only a handful of imprinted genes are known or suspected to regulate brain development (e.g., Dlk1, Peg3, Ube3a, necdin, and Grb10). Herein we show that the maternally imprinted gene Zac1 is a critical regulator of neocortical development. Our studies are relevant because loss of 6q24 maternal imprinting in humans results in elevated ZAC1 expression, which has been associated with neurocognitive defects.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Genes Supresores de Tumor/fisiología , Neocórtex/citología , Neuronas/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neocórtex/embriología , Neuritas/fisiología , Neuritas/ultraestructura , Neuronas/ultraestructura , Embarazo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Factores de Transcripción/genética
10.
Biol Psychiatry ; 78(12): 840-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25680674

RESUMEN

BACKGROUND: Posttraumatic stress disorder (PTSD) is a frequent anxiety disorder with higher prevalence rates in female patients than in male patients (2.5:1). Association with a single nucleotide polymorphism (rs2267735) in the gene ADCYAP1R1 encoding the type I receptor (PAC1-R) of the pituitary adenylate cyclase activating polypeptide has been reported with PTSD in female patients. We sought to identify the neural correlates of the described PAC1-R effects on associative learning. METHODS: In a reverse genetic approach, we examined two independent healthy samples (N1 = 112, N2 = 73) using functional magnetic resonance imaging during cued and contextual fear conditioning. Skin conductance responses and verbal self-reports of arousal, valence, and contingency were recorded. RESULTS: We found that PAC1-R modulates the blood oxygenation level-dependent response of the hippocampus. Specifically, we observed decreased hippocampal activity during contextual, but not during cued, fear conditioning in female participants carrying the PAC1-R risk allele. We observed no significant differences in conditionability for skin conductance responses, verbal reports, or activation in other brain regions between the genotype groups in female participants. CONCLUSIONS: Our results suggest that impaired contextual conditioning in the hippocampal formation may mediate the association between PAC1-R and PTSD symptoms. Our findings potentially identify a missing link between the involvement of PAC1-R in PTSD and the well-established structural and functional hippocampal deficits in these patients.


Asunto(s)
Miedo/fisiología , Hipocampo/fisiología , Polimorfismo de Nucleótido Simple , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/fisiopatología , Amígdala del Cerebelo/fisiología , Mapeo Encefálico , Condicionamiento Clásico/fisiología , Señales (Psicología) , Femenino , Respuesta Galvánica de la Piel , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Pruebas Neuropsicológicas , Factores Sexuales
11.
J Gerontol A Biol Sci Med Sci ; 69(11): 1331-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24846768

RESUMEN

With the increase of life span, nonpathological age-related memory decline is affecting an increasing number of people. However, there is evidence that age-associated memory impairment only suspends, rather than irreversibly extinguishes, the intrinsic capacity of the aging nervous system for plasticity (1). Here, using a molluscan model system, we show that the age-related decline in memory performance can be reversed by administration of the pituitary adenylate cyclase activating polypeptide (PACAP). Our earlier findings showed that a homolog of the vertebrate PACAP38 and its receptors exist in the pond snail (Lymnaea stagnalis) brain (2), and it is both necessary and instructive for memory formation after reward conditioning in young animals (3). Here we show that exogenous PACAP38 boosts memory formation in aged Lymnaea, where endogenous PACAP38 levels are low in the brain. Treatment with insulin-like growth factor-1, which in vertebrates was shown to transactivate PACAP type I (PAC1) receptors (4) also boosts memory formation in aged pond snails. Due to the evolutionarily conserved nature of these polypeptides and their established role in memory and synaptic plasticity, there is a very high probability that they could also act as "memory rejuvenating" agents in humans.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Factor I del Crecimiento Similar a la Insulina/farmacología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Lymnaea/efectos de los fármacos , Lymnaea/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Envejecimiento/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Humanos , Memoria/efectos de los fármacos , Memoria/fisiología , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Largo Plazo/fisiología , Modelos Animales , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología
12.
Exp Cell Res ; 322(1): 108-21, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24246222

RESUMEN

PACAP and its cognate peptide VIP participate in various biological functions, including myelin maturation and synthesis. However, defining whether these peptides affect peripheral expression of myelin proteins still remains unanswered. To address this issue, we assessed whether PACAP or VIP contribute to regulate the expression of three myelin proteins (MAG, MBP and MPZ, respectively) using the rat schwannoma cell line (RT4-P6D2T), a well-established model to study myelin gene expression. In addition, we endeavored to partly unravel the underlying molecular mechanisms involved. Expression of myelin-specific proteins was assessed in cells grown either in normal serum (10% FBS) or serum starved and treated with or without 100 nM PACAP or VIP. Furthermore, through pharmacological approach using the PACAP/VIP receptor antagonist (PACAP6-38) or specific pathway (MAPK or PI3K) inhibitors we defined the relative contribution of receptors and/or signaling pathways on the expression of myelin proteins. Our data show that serum starvation (24h) significantly increased both MAG, MBP and MPZ expression. Concurrently, we observed increased expression of endogenous PACAP and related receptors. Treatment with PACAP or VIP further exacerbated starvation-induced expression of myelin markers, suggesting that serum withdrawal might sensitize cells to peptide activity. Stimulation with either peptides increased phosphorylation of Akt at Ser473 residue but had no effect on phosphorylated Erk-1/2. PACAP6-38 (10 µM) impeded starvation- or peptide-induced expression of myelin markers. Similar effects were obtained after pretreatment with the PI3K inhibitor (wortmannin, 10 µM) but not the MAPKK inhibitor (PD98059, 50 µM). Together, the present finding corroborate the hypothesis that PACAP and VIP might contribute to the myelinating process preferentially via the canonical PI3K/Akt signaling pathway, providing the basis for future studies on the role of these peptides in demyelinating diseases.


Asunto(s)
Proteínas de la Mielina/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/fisiología , Células de Schwann/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología , Animales , Línea Celular Tumoral , Activación Enzimática/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Células de Schwann/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
Neuropeptides ; 47(2): 85-92, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23375386

RESUMEN

BACKGROUND: Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating peptide (PACAP)-containing nerves surround cerebral blood vessels. The peptides have potent vasodilator properties via smooth muscle cell receptors and activation of adenylate cyclase. The purpose of this study was to describe the effects of two putative VIP/PACAP receptor antagonists and the distribution of the receptor protein in rat brain vessels. METHODS: The vascular effects of VIP, PACAP-27 and PACAP-38 were investigated in segments of rat middle cerebral artery (MCA) by pressurized arteriography, and in a wire myograph. The antagonistic responses to PACAP6-38 and PG99-465 were evaluated. In addition, the receptor subtypes for VIP and PACAP (VPAC1, VPAC2 and PAC1) were visualized in the rat middle cerebral artery by immunohistochemistry and Western blotting. RESULTS: In the perfusion model, abluminal but not luminal VIP, PACAP-27 and PACAP-38 caused concentration-dependent relaxations of the MCA (27.1±0.2%, 25.2±0.4% and 0.3±0.1%, respectively). In the wire myograph, there was no significant difference in potency of the peptides in the MCA. In both systems, PACAP6-38 and PG99-465 inhibited the VIP induced relaxation. Western blot showed the presence of the receptor proteins in cerebral vasculature and immunohistochemistry showed that all three receptors are present and located in the cytoplasm of smooth muscle cells. CONCLUSION: In both systems, the two blockers antagonized the relaxant VIP effect; the potency order of agonists and the immunohistochemistry suggest the presence of the dilatory VPAC1 and VPAC2 receptors on the smooth muscle cells.


Asunto(s)
Calcio/fisiología , Arterias Cerebrales/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores de Péptido Intestinal Vasoactivo/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Angiografía , Animales , Western Blotting , Arterias Cerebrales/efectos de los fármacos , Electromiografía , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Contracción Isométrica/efectos de los fármacos , Masculino , Músculo Liso Vascular/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , 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 , Receptores de Péptido Intestinal Vasoactivo/efectos de los fármacos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Vasoconstrictores/farmacología
14.
Biol Reprod ; 88(2): 35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23197164

RESUMEN

The pituitary gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are mainly under the control of hypothalamic gonadotropin-releasing hormone (GnRH), which regulates male and female gonadal function. GnRH is released in a pulsatile manner from the hypothalamus, and the frequency of GnRH pulses determines the dominance of output of LH and FSH from pituitary gonadotrophs. That is, more rapid pulses of GnRH preferentially increase synthesis and secretion of LH, whereas FSH is preferentially stimulated by slower GnRH pulses. The detailed mechanisms underlying this phenomenon remain unknown. Pituitary adenylate cyclase-activating polypeptide (PACAP) was originally identified as a hypothalamic activator of cAMP production in pituitary cells. PACAP is produced within the pituitary gonadotroph as well as in the central nervous system. PACAP stimulates gonadotropin alpha-, LHbeta-, and FSHbeta-subunits as well as receptors for GnRH in the pituitary gonadotropin-secreting cells. In addition, its own receptor, PACAP type I receptor (PAC1R), is also regulated by PACAP in gonadotrophs. GnRH stimulates expression of PACAP as well as PAC1R, and lower frequencies of GnRH pulses preferentially increase PACAP and PAC1R expression in gonadotrophs. Increasing concentrations of PACAP further increase the levels of gonadotropin subunit and that increasing amounts of PAC1R in gonadotrophs potentiates the effects of PACAP or GnRH on gonadotropin subunit expression. In addition, we have observed that GnRH-increased FSHbeta-subunit expression was prevented in the presence of PAC1R antagonist. These observations suggest the involvement of locally produced PACAP and its PAC1R in the differential regulation of specific gonadotropin subunit expression by pulsatile GnRH stimulation. Here, we review the possible involvement of PACAP and its PAC1R in gonadotropin control on the basis of our observations with gonadotroph cell lines.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Hormona Luteinizante de Subunidad beta/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Hipófisis/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Animales , Línea Celular , Hormona Folículo Estimulante de Subunidad beta/genética , Folistatina/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Gonadotrofos/citología , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Hormona Luteinizante de Subunidad beta/genética , Ratones , Modelos Animales , Hipófisis/citología , Hipófisis/efectos de los fármacos
16.
Adv Exp Med Biol ; 758: 301-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23080176

RESUMEN

The carotid body (CB) plays important roles in cardiorespiratory changes in chronic and intermittent hypoxia. Pituitary adenylate cyclase activating polypeptide (PACAP) is involved in the regulation of respiratory chemoresponse. We hypothesized an upregulation of the expressions of PACAP and its receptor (PAC1) in the rat CB in chronic and intermittent hypoxia. The CB expressions of PACAP and PAC1 were examined in rats breathing 10% O(2) (in isobaric chamber for chronic hypoxia, 24 h/day) or in intermittent hypoxia (cyclic between air and 5% O(2) per minute, 8 h/day) for 7 days. Immunohistochemical studies showed that the PACAP and PAC1 proteins were localized in CB glomic clusters containing tyrosine hydroxylase. The proportional amount of cells with positive staining of PACAP and PAC1 was significantly increased in both hypoxic groups when compared with the normoxic control. In addition, the mRNA level of PAC1 expression was markedly elevated in the hypoxic groups, despite no changes in the PACAP expression. These results suggest an upregulation of PACAP and its receptor expression in the rat CB under chronic and intermittent hypoxic conditions. The PACAP binding to its receptor could activate the PKA signaling pathway leading to an increased CB excitability under hypoxic conditions.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Hipoxia/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Animales , Enfermedad Crónica , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/análisis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/análisis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Regulación hacia Arriba
17.
J Mol Neurosci ; 48(3): 574-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22766684

RESUMEN

Migraine is a debilitating neurological disorder characterized by mild to severe headache that is often accompanied by aura and other neurological symptoms. Among proposed mechanisms, dilation of the dural vasculature especially the middle meningeal artery (MMA) has been implicated as one component underlying this disorder. Several regulatory peptides from trigeminal sensory and sphenopalatine postganglionic parasympathetic fibers innervating these vessels have been implicated in the process including pituitary adenylate cyclase-activating polypeptide (PACAP). Although PACAP has been well described as a potent dilator in many vascular beds, the effects of PACAP on the dural vasculature are unclear. In the current study, we examined the ability of PACAP to dilate MMAs that were isolated from rats and pressurized ex vivo. PACAP38 potently dilated pressurized MMAs with an EC(50) of 1 pM. The PAC1 receptor antagonist, PACAP(6-38), abolished MMA dilation caused by picomolar concentrations of PACAP. In contrast, cerebellar arteries isolated from the brain surface were ~1,000-fold less sensitive to PACAP than MMAs. Although cerebellar arteries expressed transcripts for all three PACAP receptor subtypes (PAC1, VPAC1, and VPAC2 receptors) by RT-PCR analyses, MMA demonstrated only PAC1 and VPAC2 receptor expression. Further, multiple variants of the PAC1 receptor were identified in the MMA. The expression of PAC1 receptors and the high potency of PACAP to induce MMA vasodilation are consistent with their potential roles in the etiology of migraine.


Asunto(s)
Arterias Meníngeas/efectos de los fármacos , Trastornos Migrañosos/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Vasodilatación/efectos de los fármacos , Animales , Cerebelo/irrigación sanguínea , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Masculino , Arterias Meníngeas/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , Presión , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/biosíntesis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/biosíntesis , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/biosíntesis , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/farmacología
18.
J Mol Neurosci ; 48(3): 508-17, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22674051

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuroprotective peptide expressed in the central nervous system. To date, changes in the expression and effect of endogenous PACAP have not been clarified with respect to spinal cord injury (SCI). The aim of this study was to elucidate the expression pattern and function of endogenous PACAP on the contusion model of SCI using heterozygous PACAP knockout (PACAP(+/-)) and wild-type mice. Real-time polymerase chain reaction methods revealed that the level of PACAP mRNA increased gradually for 14 days after SCI and that PAC1R mRNA levels also increased for 7 days compared with intact control mice. PACAP and PAC1R immunoreactivities colabeled with a neuronal marker in the intact spinal cord. Seven days after SCI, PAC1R immunoreactivity was additionally co-expressed with an astrocyte marker. Wild-type mice gradually recovered motor function after 14 days, but PACAP(+/-) mice showed significantly impaired recovery from 3 days compared with wild-type mice. The injury volume at day 7 in PACAP(+/-) mice, and the number of single-stranded DNA-immunopositive cells as a marker of neuronal cell death at day 3 were significantly higher than values measured in wild-type mice. These data suggest that endogenous PACAP is upregulated by SCI and has a neuroprotective effect on the damaged spinal cord.


Asunto(s)
Proteínas del Tejido Nervioso/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Animales , Astrocitos/metabolismo , Muerte Celular , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/biosíntesis , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/deficiencia , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/biosíntesis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Recuperación de la Función , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Factores de Tiempo , Regulación hacia Arriba
19.
J Mol Neurosci ; 48(3): 526-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22678884

RESUMEN

Pituitary adenylate cyclase activating peptide (PACAP), a potent neuropeptide which crosses the blood-brain barrier, is known to provide neuroprotection in rat stroke models of middle cerebral artery occlusion (MCAO) by mechanism(s) which deserve clarification. We confirmed that following i.v. injection of 30 ng/kg of PACAP38 in rats exposed to 2 h of MCAO focal cerebral ischemia and 48 h reoxygenation, 50 % neuroprotection was measured by reduced caspase-3 activity and volume of cerebral infarction. Similar neuroprotective effects were measured upon PACAP38 treatment of oxygen-glucose deprivation and reoxygenation of brain cortical neurons. The neuroprotection was temporally associated with increased expression of brain-derived neurotrophic factor, phosphorylation of its receptor-tropomyosin-related kinase receptor type B (trkB), activation of phosphoinositide 3-kinase and Akt, and reduction of extracellular signal-regulated kinases 1/2 phosphorylation. PACAP38 increased expression of neuronal markers beta-tubulin III, microtubule-associated protein-2, and growth-associated protein-43. PACAP38 induced stimulation of Rac and suppression of Rho GTPase activities. PACAP38 downregulated the nerve growth factor receptor (p75(NTR)) and associated Nogo-(Neurite outgrowth-A) receptor. Collectively, these in vitro and in vivo results propose that PACAP exhibits neuroprotective effects in cerebral ischemia by three mechanisms: a direct one, mediated by PACAP receptors, and two indirect, induced by neurotrophin release, activation of the trkB receptors and attenuation of neuronal growth inhibitory signaling molecules p75(NTR) and Nogo receptor.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipoxia de la Célula , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Corteza Cerebral/citología , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Inyecciones Intravenosas , Masculino , Proteínas del Tejido Nervioso/genética , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , 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/fisiología , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos
20.
J Mol Neurosci ; 48(3): 660-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22581436

RESUMEN

The signal transduction mechanisms of pituitary adenylate cyclase activating polypeptide (PACAP) were investigated in lung cancer cells. Previously, PACAP-27 addition to NCI-H838 cells increased phosphatidylinositol turnover and intracellular cAMP leading to proliferation of lung cancer cells. Also, PACAP receptors (PAC1) regulated the tyrosine phosphorylation of ERK, focal adhesion kinase, and paxillin. In this communication, the effects of PACAP on cytosolic Ca(2+) and PYK-2 tyrosine phosphorylation were investigated. PACAP-27 increased cytosolic Ca(2+) within seconds after addition to FURA-2 AM loaded NCI-H838 cells. The increase in cytosolic Ca(2+) caused by PACAP was inhibited by PACAP(6-38) (PAC1 antagonist), U73122 (phospholipase C inhibitor), or BAPTA (calcium chelator), but not H89 (PKA inhibitor). PACAP-38, but not vasoactive intestinal peptide (VIP), addition to NCI-H838 or H1299 cells significantly increased the tyrosine phosphorylation of PYK-2 after 2 min. The increase in PYK-2 tyrosine phosphorylation caused by PACAP was inhibited by PACAP(6-38), U73122, or BAPTA, but not H89. The results suggest that PAC1 regulates PYK-2 tyrosine phosphorylation in a calcium-dependent manner.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Quinasa 2 de Adhesión Focal/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Señalización del Calcio/fisiología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Quelantes/farmacología , Citosol/efectos de los fármacos , Citosol/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estrenos/farmacología , Humanos , Isoquinolinas/farmacología , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirrolidinonas/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...