Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Exp Cell Res ; 423(2): 113471, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36642263

RESUMEN

Vasoactive intestinal peptide (VIP), a small neuropeptide composing of 28 amino acids, functions as a neuromodulator with insulinotropic effect on pancreatic ß cells, in which it is of vital importance in regulating the levels of blood glucose. VIP potently agonizes VPAC2 receptor (VPAC2-R). Agonists of VPAC2-R stimulate glucose-dependent insulin secretion. The purpose of this study was to further investigate the possible ion channel mechanisms in VPAC2-R-mediated VIP-potentiated insulin secretion. The results of insulin secretion experiments showed that VIP augmented insulin secretion in a glucose-dependent manner. The insulinotropic effect was mediated by VPAC2-R rather than VPAC1 receptor (VPAC1-R), through the adenylyl cyclase (AC)/protein kinase A (PKA) signalling pathway. The calcium imaging analysis demonstrated that VIP increased intracellular Ca2+ concentration ([Ca2+]i). In addition, in the whole-cell voltage-clamp mode, we found that VIP blocked the voltage-dependent potassium (Kv) channel currents, while this effect was reversed by inhibiting the VPAC2-R, AC or PKA respectively. Taken together, these findings suggest that VIP stimulates insulin secretion by inhibiting the Kv channels, activating the Ca2+ channels, and increasing [Ca2+]i through the VPAC2-R and AC/PKA signalling pathway. These findings provide theoretical basis for the research of VPAC2-R as a novel therapeutic target.


Asunto(s)
Células Secretoras de Insulina , Péptido Intestinal Vasoactivo , Ratas , Animales , Péptido Intestinal Vasoactivo/farmacología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , Glucosa/farmacología
2.
Front Endocrinol (Lausanne) ; 13: 984198, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36204104

RESUMEN

Owing to the increasing prevalence of type 2 diabetes, the development of novel hypoglycemic drugs has become a research hotspot, with the ultimate goal of developing therapeutic drugs that stimulate glucose-induced insulin secretion without inducing hypoglycemia. Vasoactive intestinal peptide (VIP), a 28-amino-acid peptide, can stimulate glucose-dependent insulin secretion, particularly by binding to VPAC2 receptors. VIP also promotes islet ß-cell proliferation through the forkhead box M1 pathway, but the specific molecular mechanism remains to be studied. The clinical application of VIP is limited because of its short half-life and wide distribution in the human body. Based on the binding properties of VIP and VPAC2 receptors, VPAC2-selective agonists have been developed to serve as novel hypoglycemic drugs. This review summarizes the physiological significance of VIP in glucose homeostasis and the potential therapeutic value of VPAC2-selective agonists in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Péptido Intestinal Vasoactivo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Secreción de Insulina , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Péptido Intestinal Vasoactivo/uso terapéutico
3.
Int J Nanomedicine ; 12: 2143-2160, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356733

RESUMEN

A novel neuroendocrine peptide, pituitary adenylate cyclase activating peptide (PACAP), was found to have an important role in carbohydrate or lipid metabolism and was susceptible to dipeptidyl peptidase IV degradation. It can not only mediate glucose-dependent insulin secretion and lower blood glucose by activating VPAC2 receptor, but also raise blood glucose by promoting glucagon production by VPAC1 receptor activation. Therefore, its therapeutic application is restricted by the exceedingly short-acting half-life and the stimulatory function for glycogenolysis. Herein, we generated novel peptide-conjugated selenium nanoparticles (SeNPs; named as SCD), comprising a 32-amino acid PACAP-derived peptide DBAYL that selectively binds to VPAC2, and chitosan-modified SeNPs (SeNPs-CTS, SC) as slow-release carrier. The circulating half-life of SCD is 14.12 h in mice, which is 168.4-and 7.1-fold longer than wild PACAP (~5 min) and DBAYL (~1.98 h), respectively. SCD (10 nmol/L) significantly promotes INS-1 cell proliferation, glucose uptake, insulin secretion, insulin receptor expression and also obviously reduces intracellular reactive oxygen species levels in H2O2-injured INS-1 cells. Furthermore, the biological effects of SCD are stronger than Exendin-4 (a clinically approved drug through its insulinotropic effect), DBAYL, SeNPs or SC. A single injection of SCD (20 nmol/kg) into db/db mice with type 2 diabetes leads to enhanced insulin secretion and sustained hypoglycemic effect, and the effectiveness and duration of SCD in enhancing insulin secretion and reducing blood glucose levels are much stronger than Exendin-4, SeNPs or SC. In db/db mice, chronic administration of SCD by daily injection for 12 weeks markedly improved glucose and lipid profiles, insulin sensitivity and the structures of pancreatic and adipose tissue. The results indicate that SC can play a role as a carrier for the slow release of bioactive peptides and SCD could be a hopeful therapeutic against type 2 diabetes through the synergy effects of DBAYL and SeNPs.


Asunto(s)
Quitosano/química , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Nanopartículas/química , Péptidos/uso terapéutico , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Selenio/química , Animales , Glucemia/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/patología , Liberación de Fármacos , Exenatida , Ayuno/sangre , Glucosa/metabolismo , Glucosa/farmacología , Semivida , Peróxido de Hidrógeno/toxicidad , Insulina/genética , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Insulina/metabolismo , Ponzoñas/uso terapéutico
4.
Neurotherapeutics ; 13(3): 635-46, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27329163

RESUMEN

Neuroprotective immunity is defined by transformation of T-cell polarity for therapeutic gain. For neurodegenerative disorders and specifically for Parkinson's disease (PD), granulocyte-macrophage colony stimulating factor or vasoactive intestinal peptide receptor 2 (VIPR2) agonists elicit robust anti-inflammatory microglial responses leading to neuronal sparing in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. While neurotherapeutic potential was demonstrated for PD, there remain inherent limitations in translating these inventions from the laboratory to patients. One obstacle in translating such novel neurotherapeutics centers on the availability of suitable noninvasive methods to track disease progression and therapeutic efficacy. To this end, we developed manganese-enhanced magnetic resonance imaging (MEMRI) assays as a way to track a linkage between glial activation and VIPR2 agonist (LBT-3627)-induced neuroprotective immunity for MPTP-induced nigrostriatal degeneration. Notably, LBT-3627-treated, MPTP-intoxicated mice show reduced MEMRI brain signal intensities. These changes paralleled reduced astrogliosis and resulted in sparing of nigral tyrosine hydroxylase neurons. Most importantly, the data suggest that MEMRI can be developed as a biomarker tool to monitor neurotherapeutic responses that are relevant to common neurodegenerative disorders used to improve disease outcomes.


Asunto(s)
Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Manganeso/administración & dosificación , Oligopéptidos/administración & dosificación , Enfermedad de Parkinson/diagnóstico por imagen , Enfermedad de Parkinson/tratamiento farmacológico , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson/inmunología , Trastornos Parkinsonianos/diagnóstico por imagen , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/inmunología , Proteínas Tirosina Quinasas/metabolismo
5.
Psychopharmacology (Berl) ; 232(12): 2181-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25575489

RESUMEN

RATIONALE: An abundance of genetic and epidemiologic evidence as well as longitudinal neuroimaging data point to developmental origins for schizophrenia and other mental health disorders. Recent clinical studies indicate that microduplications of VIPR2, encoding the vasoactive intestinal peptide (VIP) receptor VPAC2, confer significant risk for schizophrenia and autism spectrum disorder. Lymphocytes from patients with these mutations exhibited higher VIPR2 gene expression and VIP responsiveness (cAMP induction), but mechanisms by which overactive VPAC2 signaling may lead to these psychiatric disorders are unknown. OBJECTIVES: We subcutaneously administered the highly selective VPAC2 receptor agonist Ro 25-1553 to C57BL/6 mice from postnatal day 1 (P1) to P14 to determine if overactivation of VPAC2 receptor signaling during postnatal brain maturation affects synaptogenesis and selected behaviors. RESULTS: Western blot analyses on P21 revealed significant reductions of synaptophysin and postsynaptic density protein 95 (PSD-95) in the prefrontal cortex, but not in the hippocampus in Ro 25-1553-treated mice. The same postnatally restricted treatment resulted in a disruption in prepulse inhibition of the acoustic startle measured in adult mice. No effects were observed in open-field locomotor activity, sociability in the three-chamber social interaction test, or fear conditioning or extinction. CONCLUSION: Overactivation of the VPAC2 receptor in the postnatal mouse results in a reduction in synaptic proteins in the prefrontal cortex and selective alterations in prepulse inhibition. These findings suggest that the VIPR2-linkage to mental health disorders may be due in part to overactive VPAC2 receptor signaling during a critical time of synaptic maturation.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Péptidos Cíclicos/farmacología , Inhibición Prepulso/efectos de los fármacos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Homólogo 4 de la Proteína Discs Large , Miedo/efectos de los fármacos , Femenino , Guanilato-Quinasas/metabolismo , Relaciones Interpersonales , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Embarazo , Reflejo de Sobresalto/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sinaptofisina/metabolismo , Péptido Intestinal Vasoactivo/farmacología
6.
Artículo en Inglés | MEDLINE | ID: mdl-25438751

RESUMEN

Vasoactive intestinal polypeptide (VIP) is an intrinsic 28-amino acid peptide, involved in a wide range of physiologic effects, and therefore considered as a promising drug candidate for the treatment of several diseases. But the clinical application of VIP has been limited for the easy in vivo digestion. Various researches aiming to prolong the VIP half-life, by modifying the VIP structure, have been reported. The first thing to be considered after structural modification is to know it is a VPAC agonist or antagonist. To analyze the structure-activity relationships of VIP derivatives and build classifiers to distinguish newly designed VIPs, here in this work, we collected 46 samples and two classifiers were established respectively for VPAC1 and VPAC2 receptors. The built classifiers are robust and predictive with high sensitivity, specificity and concordance for the prediction set. By analyzing the meanings of the used variables, we found that the electrostatic properties of VIP derivatives are vital in their interactions with VPAC receptors. Finally, these two classifiers were used to predict the bioactivities of novel VIPs, without experimental activities, which were suggested for experimental research groups to test their bioactivities and the possible practical applications in future.


Asunto(s)
Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/antagonistas & inhibidores , Péptido Intestinal Vasoactivo/química , Péptido Intestinal Vasoactivo/farmacología , Secuencia de Aminoácidos , Simulación por Computador , Descubrimiento de Drogas , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Relación Estructura-Actividad
7.
Neuropharmacology ; 85: 538-47, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24973707

RESUMEN

We have earlier shown that PACAP-38 decreases neurogenic inflammation. However, there were no data on its receptorial mechanism and the involvement of its PAC1 and VPAC1/2 receptors (PAC1R, VPAC1/2R) in this inhibitory effect. Neurogenic inflammation in the mouse ear was induced by topical application of the Transient Receptor Potential Ankyrin 1 (TRPA1) receptor activator mustard oil (MO). Consequent neurogenic edema, vasodilation and plasma leakage were assessed by measuring ear thickness with engineer's micrometer, detecting tissue perfusion by laser Doppler scanning and Evans blue or indocyanine green extravasation by intravital videomicroscopy or fluorescence imaging, respectively. Myeloperoxidase activity, an indicator of neutrophil infiltration, was measured from the ear homogenates with spectrophotometry. The selective PAC1R agonist maxadilan, the VPAC1/2R agonist vasoactive intestinal polypeptide (VIP) or the vehicle were administered i.p. 15 min before MO. Substance P (SP) concentration of the ear was assessed by radioimmunoassay. Maxadilan significantly diminished MO-induced neurogenic edema, increase of vascular permeability and vasodilation. These inhibitory effects of maxadilan may be partially due to the decreased substance P (SP) levels. In contrast, inhibitory effect of VIP on ear swelling was moderate, without any effect on MO-induced plasma leakage or SP release, however, activation of VPAC1/2R inhibited the increased microcirculation caused by the early arteriolar vasodilation. Neither the PAC1R, nor the VPAC1/2R agonist influenced the MO-evoked increase in tissue myeloperoxidase activity. These results clearly show that PAC1R activation inhibits acute neurogenic arterial vasodilation and plasma protein leakage from the venules, while VPAC1/2R stimulation is only involved in the attenuation of vasodilation.


Asunto(s)
Proteínas de Insectos/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Fenómenos Fisiológicos de la Piel/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Modelos Animales de Enfermedad , Oído/patología , Oído/fisiopatología , Edema , Femenino , Masculino , Ratones , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Planta de la Mostaza , Peroxidasa/metabolismo , Aceites de Plantas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Sustancia P/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Vasodilatación/fisiología
8.
J Appl Physiol (1985) ; 117(4): 383-91, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24947028

RESUMEN

Pulmonary hypertension (PH) is associated with significant morbidity and mortality. Vasoactive intestinal peptide (VIP) and pituitary adenylyl cyclase activating peptide (PACAP) have pulmonary vasodilatory and positive inotropic effects via receptors VPAC1 and VPAC2, which possess a similar affinity for both peptides, and PAC1, a PACAP-preferring receptor. VIP is a promising option for PH treatment; however, various physiological effects of VIP have limited its clinical use. We investigated the effects of VPAC1 and VPAC2 selective agonists VIP and PACAP to explore more appropriate means of treatment for PH. We examined hemodynamic changes in right ventricular systolic pressure (RVSP), systemic blood pressure (SBP), total pulmonary resistance index (TPRI), total systemic resistance index, and cardiac index (CI) in response to their agonists with monocrotaline (MCT)-induced PH and explored involvement of VIP/PACAP expression and receptors in PH. Sprague-Dawley rats were divided into the MCT group (administered MCT 60 mg/kg) and control group. In MCT-induced PH, decreased VIP and PACAP were associated with upregulation of VPAC1, VPAC2, and PAC1 in lung tissues. Intravenous injection of VPAC2-selective agonist BAY 55-9837 and VIP, but not [Ala(11,22,28)]VIP, improved the CI. The decrease in SBP with VPAC2 agonist was significantly less than that in the control. Although they decreased SBP, these agonists hardly affected RVSP in the control. Activation of VPAC2 receptor with BAY 55-9837 effectively improved RVSP, TPRI, and CI in MCT-induced PH, suggesting a VPAC2 agonist as a possible promising treatment for PH.


Asunto(s)
Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/metabolismo , Monocrotalina , Venenos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Animales , Hipertrofia Ventricular Derecha/inducido químicamente , Ligandos , Masculino , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/sangre , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/sangre , Péptido Intestinal Vasoactivo/farmacología
9.
Orphanet J Rare Dis ; 9: 4, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24405637

RESUMEN

BACKGROUND: Spinal Muscular Atrophy (SMA) is one of the most common inherited causes of infant death and is caused by the loss of functional survival motor neuron (SMN) protein due to mutations or deletion in the SMN1 gene. One of the treatment strategies for SMA is to induce the expression of the protein from the homologous SMN2 gene, a rescuing paralog for SMA. METHODS AND RESULTS: Here we demonstrate the promise of pharmacological modulation of SMN2 gene by BAY 55-9837, an agonist of the vasoactive intestinal peptide receptor 2 (VPAC2), a member of G protein coupled receptor family. Treatment with BAY 55-9837 lead to induction of SMN protein levels via activation of MAPK14 or p38 pathway in vitro. Importantly, BAY 55-9837 also ameliorated disease phenotype in severe SMA mouse models. CONCLUSION: Our findings suggest the VPAC2 pathway is a potential SMA therapeutic target.


Asunto(s)
Atrofia Muscular Espinal/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Péptido Intestinal Vasoactivo/uso terapéutico
10.
Eur J Pharmacol ; 713(1-3): 25-30, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23665113

RESUMEN

PACAP and VIP have prominent effects on cardiac function in several species, but little is known about their influence on the murine heart. Accordingly, we evaluated the expression of PACAP/VIP receptors in mouse heart and the response of isolated atria to peptide agonists. Quantitative PCR demonstrated that PAC1, VPAC1, and VPAC2 receptor mRNAs are present throughout the mouse heart. Expression of all three receptor transcripts was low, PAC1 being the lowest. No regional differences in expression were detected for individual receptor mRNAs after normalization to L32. Pharmacological effects of PACAP-27, VIP, and the selective PAC1 agonist maxadilan were evaluated in isolated, spontaneously beating atria from C57BL/6 mice of either sex. Incremental additions of PACAP-27 at 1 min intervals caused a concentration-dependent tachycardia with a logEC50=-9.08 ± 0.15 M (n=7) and a maximum of 96.3 ± 5.9% above baseline heart rate. VIP and maxadilan also caused tachycardia but their potencies were about two orders of magnitude less. Increasing the dosing interval to 5 min caused a leftward shift of the concentration-response curve to maxadilan but no changes in the curves for PACAP-27 or VIP. Under this condition, neither the potency nor the efficacy of maxadilan differed from those of PACAP-27. Neither PACAP-27 nor maxadilan caused tachyphylaxis, and maximal responses to maxadilan were maintained for at least 2 h. We conclude that all three VIP/PACAP family receptors are expressed by mouse cardiac tissue, but only PAC1 receptors mediate positive chronotropic responses to PACAP-27 and VIP.


Asunto(s)
Atrios Cardíacos/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Atrios Cardíacos/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Especificidad de la Especie
11.
Invest Ophthalmol Vis Sci ; 54(4): 2872-84, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23518767

RESUMEN

PURPOSE: To determine the intracellular signaling pathways that vasoactive intestinal peptide (VIP) uses to stimulate high molecular weight glycoconjugate secretion from cultured rat conjunctival goblet cells. METHODS: Goblet cells from rat bulbar and forniceal conjunctiva were grown in organ culture. Presence and localization of VIP receptors (VPAC1 and 2) were determined by RT-PCR, immunofluorescence microscopy and Western blot analysis. Intracellular [Ca(2+)] ([Ca(2+)]i) was measured using fura-2. Extracellular signal-regulated kinase (ERK)-1/2 activity was determined by Western blot analysis. High molecular weight glycoconjugate secretion was measured with an enzyme-linked lectin assay on cultured goblet cells that were serum-starved for 2 hours before stimulation with VIP, VPAC1-, or VPAC2-specific agonists. Inhibitors were added 30 minutes prior to VIP. Activation of epidermal growth factor receptor (EGFR) was measured by immunoprecipitation using an antibody against pTyr followed by Western blot analysis with an antibody against EGFR. RESULTS: Both VIP receptors were present in rat conjunctiva and cultured goblet cells. VIP- and VPAC-specific agonists increased [Ca(2+)]i and secretion in a concentration-dependent manner. VIP also increased ERK1/2 activity, VIP-stimulated increase in [Ca(2+)]i. Secretion, but not ERK1/2 activity, was inhibited by the protein kinase A inhibitor, H89. VIP-stimulated secretion was inhibited by siRNA for ERK2 but not by siRNA for EGFR. VIP did not increase the phosphorylation of the EGFR. CONCLUSIONS: In conclusion, in cultured rat conjunctival goblet cells, VPAC1 and 2 receptors are functional. VIP stimulates a cAMP-dependent increase in [Ca(2+)]i and glycoconjugate secretion, but not ERK1/2 activation. VIP does not activate with EGFR.


Asunto(s)
Conjuntiva/metabolismo , Células Caliciformes/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Conjuntiva/citología , Conjuntiva/inervación , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN Complementario/genética , Glicoconjugados/metabolismo , Células Caliciformes/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Mucina 5AC/metabolismo , Técnicas de Cultivo de Órganos , Sistema Nervioso Parasimpático/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/farmacología
12.
Neuropeptides ; 47(2): 133-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22981158

RESUMEN

VPAC2 receptor is a potential target for the treatment of type 2 diabetes and may also convey neuroprotective effects. The aim of this study was to determine the potential efficacy of the VPAC2 receptor agonist Bay 55-9837 against stroke in type-2 diabetic Goto-Kakizaki (GK) rats. GK rats were treated intravenously once daily for 7 days with 0.25 or 0.025 nmol/kg Bay 55-9837 or vehicle before inducing stroke by transient middle cerebral artery occlusion. Treatments were then continued for 7 further days. The glycemic effects of Bay 55-9837 were assessed by measuring fasting blood glucose and oral glucose tolerance. The severity of stroke was measured by assessing ischemic volume. The results show that Bay 55-9837 is not effective in lowering fasting glycemia and does not facilitate glucose disposal. The highest dose of Bay 55-9837 (0.25 nmol/kg) led to increased mortality and brain hemorrhage when compared to control. The lower dose of Bay 55-9837 (0.025 nmol/kg) did not increase mortality rate but caused a threefold increase of the ischemic lesion size with signs of brain hemorrhages as compared to control. In conclusion, Bay 55-9837 did not show antidiabetic or antistroke efficacy in the type 2 diabetic GK rat. Contrarily, Bay 55-9837 treatment led to increased mortality and worsening of the severity of stroke.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Hemorragias Intracraneales/patología , Neuronas/patología , Fragmentos de Péptidos/farmacología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Accidente Cerebrovascular/patología , Animales , Glucemia , Recuento de Células , Progresión de la Enfermedad , Prueba de Tolerancia a la Glucosa , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Masculino , Arteria Cerebral Media/fisiología , Neuronas/efectos de los fármacos , Ratas , Péptido Intestinal Vasoactivo/farmacología
13.
Cephalalgia ; 31(2): 181-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20974589

RESUMEN

OBJECTIVE: We pharmacologically characterized pituitary adenylate cyclase-activating polypeptides (PACAPs), vasoactive intestinal peptide (VIP) and the VPAC(1), VPAC(2) and PAC(1) receptors in human meningeal (for their role in migraine) and coronary (for potential side effects) arteries. METHODS: Concentration response curves to PACAP38, PACAP27, VIP and the VPAC(1) receptor agonist ([Lys15,Arg16,Leu27]-VIP[1-7]-GRF[8-27]) were constructed in the absence or presence of the PAC(1) receptor antagonist PACAP6-38 or the VPAC(1) receptor antagonist, PG97269. mRNA expression was measured using qPCR. RESULTS: PACAP38 was less potent than VIP in both arteries. Both peptides had lower potency and efficacy in meningeal than in coronary arteries, while mRNA expression of VPAC(1) receptor was more pronounced in meningeal arteries. PACAP6-38 reduced the E(max) of PACAP27, while PG97269 right-shifted the VIP-induced relaxation curve only in the coronary arteries. CONCLUSION: The direct vasodilatory effect of VIP and PACAP might be less relevant than the central effect of this compound in migraine pathogenesis.


Asunto(s)
Vasos Coronarios/fisiología , Arterias Meníngeas/fisiología , Trastornos Migrañosos/fisiopatología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Receptores de Péptido Intestinal Vasoactivo , Adulto , Anciano , Vasos Coronarios/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Masculino , Arterias Meníngeas/efectos de los fármacos , Persona de Mediana Edad , Trastornos Migrañosos/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/efectos adversos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/antagonistas & inhibidores , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , ARN Mensajero/metabolismo , 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/genética , Receptores de Péptido Intestinal Vasoactivo/agonistas , Receptores de Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Receptores de Péptido Intestinal Vasoactivo/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/antagonistas & inhibidores , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/análogos & derivados , Péptido Intestinal Vasoactivo/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
14.
Biochem Biophys Res Commun ; 402(3): 471-6, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20951679

RESUMEN

OBJECTIVE: Vasoactive intestinal peptide (VIP) is a 28-amino acid peptide widely expressed in the body and binding three types of receptors: VPAC(1)-R, VPAC(2)-R and PAC(1)-R. Based on beneficial effects of VIP and VPAC(1)-R agonists in mouse models of several chronic inflammatory disorders, we hypothesized that activation of VIP receptors would prevent atherosclerosis development in apolipoprotein E-deficient mice. METHODS AND RESULTS: Contrary to our hypothesis, administration of a VPAC(1)-R agonist, (Ala(11,22,28))-VIP aggravated atherosclerotic lesion development in the aortic root of these mice compared to control mice. This was accompanied by a significant increase in the expression of MHC class II protein I-A(b), and suggests enhanced inflammatory activity in the vessel wall. The amount of macrophage-specific CD68 staining as well as serum cholesterol and triglyceride levels did not change as a result of the (Ala(11,22,28))-VIP treatment, i.e. the treatment resulted in significant changes in lipid accumulation in the lesions without changing the number of macrophages or systemic lipid levels. Interestingly, administration of VIP did not alter the course of the disease. CONCLUSION: Despite beneficial effects in murine models of several inflammatory disorders, VPAC(1)-R activation aggravates atherosclerotic lesion formation in apolipoprotein E-deficient mice through enhanced inflammatory activity in the vessel wall.


Asunto(s)
Aterosclerosis/patología , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Animales , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Linfocitos B/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Inflamación/etiología , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Noqueados , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Péptido Intestinal Vasoactivo/sangre , Péptido Intestinal Vasoactivo/farmacología
15.
Br J Pharmacol ; 161(6): 1329-42, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20735404

RESUMEN

BACKGROUND AND PURPOSE: Vasoactive intestinal peptide is expressed in the respiratory tract and induces its effects via its receptors, VPAC(1) and VPAC(2). RO5024118 is a selective VPAC(2) receptor agonist derived via chemical modification of an earlier VPAC(2) agonist, RO0251553. In the present studies, we characterized the pharmacological activity of RO5024118. EXPERIMENTAL APPROACH: Stability of RO5024118 to human neutrophil elastase was assessed. Bronchodilatory activity of RO5024118 was investigated in guinea pig and human isolated airway smooth muscle preparations and in a guinea pig bronchoconstriction model. Pulmonary anti-inflammatory activity of RO5024118 was investigated in a lipopolysaccharide mouse model and in a porcine pancreatic elastase (PPE) rat model. KEY RESULTS: RO5024118 demonstrated increased stability to neutrophil elastase compared with RO0251553. In human and guinea pig isolated airway preparations, RO5024118 induced bronchodilatory effects comparable with RO0251553 and the long-acting ß-agonist salmeterol and was significantly more potent than native vasoactive intestinal peptide and the short-acting ß-agonist salbutamol. In 5-HT-induced bronchoconstriction in guinea pigs, RO5024118 exhibited inhibitory activity with similar efficacy as, and longer duration than, RO0251553. In a lipopolysaccharide-mouse model, RO5024118 inhibited neutrophil and CD8(+) cells and myeloperoxidase levels. In rats, intratracheal instillation of PPE induced airway neutrophilia that was resistant to dexamethasone. Pretreatment with RO5024118 significantly inhibited PPE-induced neutrophil accumulation. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that RO5024118 induces dual bronchodilatory and pulmonary anti-inflammatory activity and may be beneficial in treating airway obstructive and inflammatory diseases. LINKED ARTICLES: This article is part of a themed section on Analytical Receptor Pharmacology in Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2010.161.issue-6.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Broncoconstricción/efectos de los fármacos , Broncodilatadores/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Péptido Intestinal Vasoactivo/farmacología , Secuencia de Aminoácidos , Animales , Antiinflamatorios no Esteroideos/metabolismo , Broncoconstricción/fisiología , Broncodilatadores/metabolismo , Cobayas , Células HT29 , Humanos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ratas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Porcinos , Péptido Intestinal Vasoactivo/agonistas , Péptido Intestinal Vasoactivo/metabolismo
16.
Acta Biochim Biophys Sin (Shanghai) ; 42(1): 21-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20043043

RESUMEN

The development of rBAY, a recombinant peptide with the similar sequence of synthetic BAY55-9837, as a potential peptide therapeutic for type 2 diabetes is still a challenge mainly because of its poor stability in aqueous solution. To improve the peptide stability and bioactivity and investigate its biological effects for VPAC2-specific activation, RBAYL with 31 aa was designed based on sequence alignments of pituitary adenylate cyclase-activating peptides (PACAPs), vasoactive intestinal peptide (VIP), and related analogs and generated through sitedirected mutagenesis. Stability analysis showed that the prepared RBAYL with three mutations (N9Q, V17L, and N28K) were much more stable than rBAY. rRBAYL (the recombinant RBAYL) was expressed and purified by gene-recombination technology via native thiol ligation on solid beads. As much as 27.7 mg rRBAYL peptide with purity over 98% was obtained from 1 L of LB medium without expensive high-performance liquid chromatography refinements. The bioactivity assay of rRBAYL showed that it displaced [(125)I]PACAP38 and [(125)I]VIP from VPAC2 with a half-maximal inhibitory concentration of 51+/-6 and 50+/-4 nM, respectively, which were similar to those of the chemically synthesized RBAYL (sRBAYL) and lower than those of Ro25-1553, an established VPAC2 agonist. rRBAYL enhances the cAMP accumulation in CHO cells expressing human VPAC2 with a half-maximal stimulatory concentration (EC50) of 0.91 nM, whereas the receptor potency of rRBAYL at human VPAC1 (EC50 of 719 nM) was only 1/790 of that at human VPAC2, and rRBAYL had no activity toward human PAC1 receptor. Western-blot assay for glucose transporter 4 (GLUT4) indicated that the rRBAYL could significantly induce GLUT4 expression more efficiently than rBAY or Ro25-1553 in adipocytes. Compared with rBAY, rRBAYL can more efficiently promote insulin release and decrease plasma glucose level in ICR mice. Our results suggested that rRBAYL is a novel recombinant VPAC2-specific agonist with high stability and bioactivity.


Asunto(s)
Adipocitos/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Transducción de Señal/efectos de los fármacos , Adipocitos/metabolismo , Animales , Unión Competitiva , Glucemia , Células CHO , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos ICR , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
17.
Stem Cells ; 27(10): 2539-51, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19650041

RESUMEN

The controlled production of neurons in the postnatal dentate gyrus and throughout life is important for hippocampal learning and memory. The mechanisms underlying the necessary coupling of neuronal activity to neural stem/progenitor cell (NSPC) function remain poorly understood. Within the dentate subgranular stem cell niche, local interneurons appear to play an important part in this excitation-neurogenesis coupling via GABAergic transmission, which promotes neuronal differentiation and integration. Here we show that vasoactive intestinal polypeptide, a neuropeptide coreleased with GABA under specific firing conditions, is uniquely trophic for proliferating postnatal nestin-positive dentate NSPCs, mediated via the VPAC(2) receptor. We also show that VPAC(2) receptor activation shifts the fate of symmetrically dividing NSPCs toward a nestin-only phenotype, independent of the trophic effect. In contrast, selective VPAC(1) receptor activation shifts NSPC fate toward granule cell neurogenesis without any trophism. We confirm a trophic role for VPAC(2) receptors in vivo, showing reduced progeny survival and dentate neurogenesis in adult Vipr2(-/-) mice. We also show a specific reduction in type 2 nestin-positive precursors in vivo, consistent with a role for VPAC(2) in maintaining this cell population. This work provides the first evidence of differential fate modulation of neurogenesis by neurotransmitter receptor subtypes and extends the fate-determining effects of neurotransmitters to maintaining the nestin-positive pool of NSPCs. This differential receptor effect may support the independent pharmacological manipulation of precursor pool expansion and neurogenic instruction for therapeutic application in the treatment of cognitive deficits associated with a decline in neurogenesis.


Asunto(s)
Giro Dentado/crecimiento & desarrollo , Neuronas/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Células Madre/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Giro Dentado/citología , Femenino , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neuronas/citología , Neuronas/efectos de los fármacos , Fenotipo , Ratas , Ratas Wistar , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Células Madre/citología , Células Madre/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología
18.
Hippocampus ; 19(1): 99-109, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18727050

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP), a neurotrophic and neuromodulatory peptide, was recently shown to enhance NMDA receptor-mediated currents in the hippocampus (Macdonald, et al. 2005. J Neurosci 25:11374-11384). To check if PACAP might also modulate AMPA receptor function, we tested its effects on AMPA receptor-mediated synaptic currents on CA1 pyramidal neurons, using the patch clamp technique on hippocampal slices. In the presence of the NMDA antagonist D-AP5, PACAP (10 nM) reduced the amplitude of excitatory postsynaptic currents (EPSCs) evoked in CA1 pyramidal neurons by stimulation of Schaffer collaterals. Following a paired-pulse stimulation protocol, the paired-pulse ratio was unaffected in most neurons, suggesting that the AMPA-mediated EPSC was modulated by PACAP mainly at a postsynaptic level. PACAP also modulated the currents induced on CA1 pyramidal neurons by applications of either glutamate or AMPA. The effects of PACAP were dose-dependent: at a 0.5 nM dose, PACAP increased AMPA-mediated current; such effect was blocked by PACAP 6-38, a selective antagonist of PAC1 receptors. The enhancement of AMPA-mediated current by PACAP 0.5 nM was abolished when cAMPS-Rp, a PKA inhibitor, was added to the intracellular solution. At a 10 nM concentration, PACAP reduced AMPA-mediated current; such effect was not blocked by PACAP 6-38. The inhibitory effect of 10 nM PACAP was mimicked by Bay 55-9837 (a selective agonist of VPAC2 receptors), persisted in the presence of intracellular BAPTA and was abolished by intracellular cAMPS-Rp. Stimulation-evoked EPSCs in CA1 neurons were significantly reduced following application of the PAC1 antagonist PACAP 6-38; this result indicates that PAC1 receptors in the CA1 region are tonically activated by endogenous PACAP and enhance CA3-CA1 synaptic transmission. Our results show that PACAP differentially modulates AMPA receptor-mediated current in CA1 pyramidal neurons by activation of PAC1 and VPAC2 receptors, both involving the cAMP/PKA pathway; the functional significance will be discussed in light of the multiple effects exerted by PACAP on the CA3-CA1 synapse at different levels.


Asunto(s)
Hipocampo/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Células Piramidales/metabolismo , Receptores AMPA/metabolismo , Potenciales Sinápticos/fisiología , Transmisión Sináptica/fisiología , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Receptores AMPA/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Potenciales Sinápticos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
19.
Pharmacol Ther ; 121(3): 294-316, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19109992

RESUMEN

Vasoactive intestinal peptide (VIP) and the pituitary adenylate cyclase activating polypeptides (PACAPs) share 68% identity at the amino acid level and belong to the secretin peptide family. Following the initial discovery of VIP almost four decades ago a substantial amount of knowledge has been presented describing the mechanisms of action, distribution and pleiotropic functions of these related peptides. It is now known that the physiological actions of these widely distributed peptides are produced through activation of three common G-protein coupled receptors (VPAC(1), VPAC(2) and PAC(1)R) which preferentially stimulate adenylate cyclase and increase intracellular cAMP, although stimulation of other intracellular messengers, including calcium and phospholipase D, has been reported. Using a range of in vitro and in vivo approaches, including cell-based functional assays, transgenic animals and rodent models of disease, VPAC/PAC receptor activation has been associated with numerous physiological processes (e.g. control of circadian rhythms) and clinical conditions (e.g. pulmonary hypertension), which underlies on-going research efforts and makes these peptides and their cognate receptors attractive targets for the pharmaceutical industry. However, despite the considerable interest in VPAC/PAC receptors and the processes which they mediate, there is still a paucity of selective and available, non-peptide ligands, which has hindered further advances in this field both at the basic research and clinical level. This review summarises the current knowledge of VIP/PACAP and the VPAC/PAC receptors with regard to their distribution, pharmacology, signalling pathways, splice variants and finally, the utility of animal models in exploring their physiological roles.


Asunto(s)
Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/fisiología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/fisiología , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , AMP Cíclico/metabolismo , Ligandos , Ratones , Ratones Transgénicos , Especificidad de Órganos , Fosfolipasa D/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Transducción de Señal , Péptido Intestinal Vasoactivo/fisiología
20.
Ann N Y Acad Sci ; 1144: 28-35, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19076360

RESUMEN

In the pancreatic islets, pituitary adenylate cyclase-activating polypeptide (PACAP) is expressed in beta cells and autonomic nerve terminals; the majority of these nerve terminals are parasympathetic. PACAP binds to three types of G protein-coupled receptors (GPCRs): VPAC1 receptors, VPAC2 receptors, and PAC1 receptors. All these receptor types are expressed in pancreatic islets. PACAP stimulates insulin and glucagon secretion. These actions are achieved in part through increased formation of cAMP after activation of adenylate cyclase and in part through increase in cytosolic calcium, achieved through increase in calcium uptake and release from intracellular calcium stores. Deletion of PAC1 receptors or VPAC2 receptors results in impaired insulin secretion and glucose intolerance. Studies in PAC1 receptor gene deleted mice have suggested that PACAP may be of physiological importance in mediating prandial insulin secretion and in contributing to the glucagon response to hypoglycemia. Animal studies have also suggested that activation of the receptors, in particular VPAC2 receptors, may be used as a therapeutic approach for the treatment of type 2 diabetes. Hence, PACAP is an islet neuropeptide with a potential role in islet physiology and as a basis for development of islet-promoting therapy in type 2 diabetes.


Asunto(s)
Páncreas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Sistema Endocrino/metabolismo , Glucagón/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...