Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 12: 711906, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867774

RESUMO

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two neuropeptides that contribute to the regulation of intestinal motility and secretion, exocrine and endocrine secretions, and homeostasis of the immune system. Their biological effects are mediated by three receptors named VPAC1, VPAC2 and PAC1 that belong to class B GPCRs. VIP and PACAP receptors have been identified as potential therapeutic targets for the treatment of chronic inflammation, neurodegenerative diseases and cancer. However, pharmacological use of endogenous ligands for these receptors is limited by their lack of specificity (PACAP binds with high affinity to VPAC1, VPAC2 and PAC1 receptors while VIP recognizes both VPAC1 and VPAC2 receptors), their poor oral bioavailability (VIP and PACAP are 27- to 38-amino acid peptides) and their short half-life. Therefore, the development of non-peptidic small molecules or specific stabilized peptidic ligands is of high interest. Structural similarities between VIP and PACAP receptors are major causes of difficulties in the design of efficient and selective compounds that could be used as therapeutics. In this study we performed structure-based virtual screening against the subset of the ZINC15 drug library. This drug repositioning screen provided new applications for a known drug: ticagrelor, a P2Y12 purinergic receptor antagonist. Ticagrelor inhibits both VPAC1 and VPAC2 receptors which was confirmed in VIP-binding and calcium mobilization assays. A following analysis of detailed ticagrelor binding modes to all three VIP and PACAP receptors with molecular dynamics revealed its allosteric mechanism of action. Using a validated homology model of inactive VPAC1 and a recently released cryo-EM structure of active VPAC1 we described how ticagrelor could block conformational changes in the region of 'tyrosine toggle switch' required for the receptor activation. We also discuss possible modifications of ticagrelor comparing other P2Y12 antagonist - cangrelor, closely related to ticagrelor but not active for VPAC1/VPAC2. This comparison with inactive cangrelor could lead to further improvement of the ticagrelor activity and selectivity for VIP and PACAP receptor sub-types.


Assuntos
Regulação Alostérica/efeitos dos fármacos , Reposicionamento de Medicamentos/métodos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Ticagrelor/farmacologia , Sítios de Ligação , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos/métodos , Estrutura Molecular , Conformação Proteica/efeitos dos fármacos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/química , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/química , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/química , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Ticagrelor/química
2.
Molecules ; 26(16)2021 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-34443519

RESUMO

The search for efficacious treatment of neurodegenerative and progressive neuroinflammatory diseases continues, as current therapies are unable to halt or reverse disease progression. PACAP represents one potential therapeutic that provides neuroprotection effects on neurons, and also modulates inflammatory responses and circulation within the brain. However, PACAP is a relatively long peptide hormone that is not trivial to synthesize. Based on previous observations that the shortened isoform PACAP1-23 is capable of inducing neuroprotection in vitro, we were inspired to synthesize shortened glycopeptide analogues of PACAP1-23. Herein, we report the synthesis and in vitro characterization of glycosylated PACAP1-23 analogues that interact strongly with the PAC1 and VPAC1 receptors, while showing reduced activity at the VPAC2 receptor.


Assuntos
Glicopeptídeos/química , Inflamação/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fragmentos de Peptídeos/química , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Glicopeptídeos/síntese química , Glicopeptídeos/farmacologia , Humanos , Inflamação/patologia , Doenças Neurodegenerativas/patologia , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Hormônios Peptídicos/síntese química , Hormônios Peptídicos/química , Hormônios Peptídicos/farmacologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos
3.
Neurobiol Learn Mem ; 180: 107423, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33705861

RESUMO

Social recognition memory (SRM) forms the basis of social relationships of animals. It is essential for social interaction and adaptive behavior, reproduction and species survival. Evidence demonstrates that social deficits of psychiatric disorders such as autism and schizophrenia are caused by alterations in SRM processing by the hippocampus and amygdala. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and its receptors PAC1, VPAC1 and VPAC2 are highly expressed in these regions. PACAP is a pleiotropic neuropeptide that modulates synaptic function and plasticity and is thought to be involved in social behavior. PACAP signaling also stimulates the nitric oxide (NO) production and targets outcomes to synapses. In the present work, we investigate the effect of the infusion of PACAP-38 (endogenous neuropeptide and potent stimulator of adenylyl cyclase), PACAP 6-38 (PAC1/VPAC2 receptors antagonist) and S-Nitroso-N-acetyl-DL-penicillamine (SNAP, NO donor) in the CA1 region of the hippocampus and in the basolateral amygdala (BLA) on the consolidation of SRM. For this, male Wistar rats with cannulae implanted in CA1 or in BLA were subjected to a social discrimination paradigm, which is based on the natural ability of rodents to investigate unfamiliar conspecifics more than familiar one. In the sample phase (acquisition), animals were exposed to a juvenile conspecific for 1 h. Immediately, 60 or 150 min after, animals received one of different pharmacological treatments. Twenty-four hours later, they were submitted to a 5 min retention test in the presence of the previously presented juvenile (familiar) and a novel juvenile. Animals that received infusions of PACAP 6-38 (40 pg/side) into CA1 immediately after the sample phase or into BLA immediately or 60 min after the sample phase were unable to recognize the familiar juvenile during the retention test. This impairment was abolished by the coinfusion of PACAP 6-38 plus SNAP (5 µg/side). These results show that the blockade of PACAP/PAC1/VPAC2 signaling in the CA1 and BLA during a restricted post-acquisition time window impairs the consolidation of SRM and that the SNAP is able to abolish this deficit. Findings like this could potentially be used in the future to influence studies of psychiatric disorders involving social behavior.


Assuntos
Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Percepção Social/efeitos dos fármacos , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Região CA1 Hipocampal/metabolismo , Consolidação da Memória/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Reconhecimento Psicológico/fisiologia , S-Nitroso-N-Acetilpenicilamina/farmacologia
4.
J Leukoc Biol ; 100(6): 1385-1393, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27381006

RESUMO

This study tested the hypothesis that vasoactive intestinal peptide (VIP) is able to modify the macrophage inflammatory profile, thus supporting its therapeutic role in autoimmune diseases. Macrophages are innate immune cells that display a variety of functions and inflammatory profiles in response to the environment that critically controls their polarization. Deregulation between the pro- and anti-inflammatory phenotypes has been involved in different pathologies. Rheumatoid arthritis (RA) is an autoimmune disease, in which macrophages are considered central effectors of synovial inflammation, displaying a proinflammatory profile. VIP is a pleiotropic neuropeptide with proven anti-inflammatory actions. As modulation of the macrophage phenotype has been implicated in the resolution of inflammatory diseases, we evaluated whether VIP is able to modulate human macrophage polarization. In vitro-polarized macrophages by GM-CSF (GM-MØ), with a proinflammatory profile, expressed higher levels of VIP receptors, vasoactive intestinal polypeptide receptors 1 and 2 (VPAC1 and VPAC2, respectively), than macrophages polarized by M-CSF (M-MØ) with anti-inflammatory activities. RA synovial macrophages, according to their GM-CSF-like polarization state, expressed both VPAC1 and VPAC2. In vitro-generated GM-MØ exposed to VIP exhibited an up-regulation of M-MØ gene marker expression, whereas their proinflammatory cytokine profile was reduced in favor of an anti-inflammatory function. Likewise, in GM-MØ, generated in the presence of VIP, VIP somehow changes the macrophages physiology profile to a less-damaging phenotype. Therefore, these results add new value to VIP as an immunomodulatory agent on inflammatory diseases.


Assuntos
Macrófagos/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/fisiologia , Peptídeo Intestinal Vasoativo/farmacologia , Artrite Reumatoide/patologia , Células Cultivadas , AMP Cíclico/metabolismo , Citocinas/biossíntese , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Imunofenotipagem , Inflamação , Ativação de Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/metabolismo , RNA Mensageiro/biossíntese , Receptores Tipo II de Peptídeo Intestinal Vasoativo/biossíntese , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/biossíntese , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Líquido Sinovial/citologia , Regulação para Cima
5.
J Neurosci ; 35(50): 16463-78, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26674871

RESUMO

Vasoactive intestinal peptide (VIP) mediates a broad range of biological responses by activating two related receptors, VIP receptor 1 and 2 (VIPR1 and VIPR2). Although the use of native VIP facilitates neuroprotection, clinical application of the hormone is limited due to VIP's rapid metabolism and inability to distinguish between VIPR1 and VIPR2 receptors. In addition, activation of both receptors by therapeutics may increase adverse secondary toxicities. Therefore, we developed metabolically stable and receptor-selective agonists for VIPR1 and VIPR2 to improve pharmacokinetic and pharmacodynamic therapeutic end points. Selective agonists were investigated for their abilities to protect mice against MPTP-induced neurodegeneration used to model Parkinson's disease (PD). Survival of tyrosine hydroxylase neurons in the substantia nigra was determined by stereological tests after MPTP intoxication in mice pretreated with either VIPR1 or VIPR2 agonist or after adoptive transfer of splenic cell populations from agonist-treated mice administered to MPTP-intoxicated animals. Treatment with VIPR2 agonist or splenocytes from agonist-treated mice resulted in increased neuronal sparing. Immunohistochemical tests showed that agonist-treated mice displayed reductions in microglial responses, with the most pronounced effects in VIPR2 agonist-treated, MPTP-intoxicated mice. In parallel studies, we observed reductions in proinflammatory cytokine release that included IL-17A, IL-6, and IFN-γ and increases in GM-CSF transcripts in CD4(+) T cells recovered from VIPR2 agonist-treated animals. Moreover, a phenotypic shift of effector to regulatory T cells was observed. These results support the use of VIPR2-selective agonists as neuroprotective agents for PD treatment. SIGNIFICANCE STATEMENT: Vasoactive intestinal peptide receptor 2 can elicit immune transformation in a model of Parkinson's disease (PD). Such immunomodulatory capabilities can lead to neuroprotection by attenuating microglial activation and by slowing degradation of neuronal cell bodies and termini in MPTP-intoxicated mice. The protective mechanism arises from altering a Th1/Th2 immune cytokine response into an anti-inflammatory and neuronal sparing profile. These results are directly applicable for the development of novel PD therapies.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/imunologia , Intoxicação por MPTP/tratamento farmacológico , Intoxicação por MPTP/imunologia , Fármacos Neuroprotetores/uso terapêutico , Oligopeptídeos/farmacologia , Receptores de Peptídeo Intestinal Vasoativo/agonistas , Animais , Linfócitos T CD4-Positivos/metabolismo , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Citocinas/metabolismo , Humanos , Imuno-Histoquímica , Intoxicação por MPTP/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Oligopeptídeos/farmacocinética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Baço/citologia , Baço/efeitos dos fármacos , Substância Negra/enzimologia , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G748-58, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24578344

RESUMO

In the gastrointestinal tract, vasoactive intestinal peptide (VIP) is found exclusively within neurons. VIP regulates intestinal motility via neurally mediated and direct actions on smooth muscle and secretion by a direct mucosal action, and via actions on submucosal neurons. VIP acts via VPAC1 and VPAC2 receptors; however, the subtype involved in its neural actions is unclear. The neural roles of VIP and VPAC1 receptors (VPAC1R) were investigated in intestinal motility and secretion in guinea pig jejunum. Expression of VIP receptors across the jejunal layers was examined using RT-PCR. Submucosal and myenteric neurons expressing VIP receptor subtype VPAC1 and/or various neurochemical markers were identified immunohistochemically. Isotonic muscle contraction was measured in longitudinal muscle-myenteric plexus preparations. Electrogenic secretion across mucosa-submucosa preparations was measured in Ussing chambers by monitoring short-circuit current. Calretinin(+) excitatory longitudinal muscle motor neurons expressed VPAC1R. Most cholinergic submucosal neurons, notably NPY(+) secretomotor neurons, expressed VPAC1R. VIP (100 nM) induced longitudinal muscle contraction that was inhibited by TTX (1 µM), PG97-269 (VPAC1 antagonist; 1 µM), and hyoscine (10 µM), but not by hexamethonium (200 µM). VIP (50 nM)-evoked secretion was depressed by hyoscine or PG97-269 and involved a small TTX-sensitive component. PG97-269 and TTX combined did not further depress the VIP response observed in the presence of PG97-269 alone. We conclude that VIP stimulates ACh-mediated longitudinal muscle contraction via VPAC1R on cholinergic motor neurons. VIP induces Cl(-) secretion directly via epithelial VPAC1R and indirectly via VPAC1R on cholinergic secretomotor neurons. No evidence was obtained for involvement of other neural VIP receptors.


Assuntos
Neurônios Colinérgicos/metabolismo , Motilidade Gastrointestinal , Secreções Intestinais/metabolismo , Jejuno/inervação , Jejuno/metabolismo , Contração Muscular , Músculo Liso/inervação , Músculo Liso/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Acetilcolina/metabolismo , Animais , Calbindina 2/metabolismo , Cloretos/metabolismo , Neurônios Colinérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Motilidade Gastrointestinal/efeitos dos fármacos , Cobaias , Mucosa Intestinal/inervação , Mucosa Intestinal/metabolismo , Jejuno/efeitos dos fármacos , Masculino , Potenciais da Membrana , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Neurotransmissores/farmacologia , RNA Mensageiro/metabolismo , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Transdução de Sinais , Peptídeo Intestinal Vasoativo/metabolismo
7.
Invest Ophthalmol Vis Sci ; 53(12): 7432-9, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23036997

RESUMO

PURPOSE: Studies from our laboratory have demonstrated that vasoactive intestinal peptide (VIP) directly converts the normally susceptible C57BL/6J (B6) mouse to resistant after ocular infection through modulation of the inflammatory response. This study examines mechanisms by which VIP influences the healing phase following infection--specifically reconstitution of the extracellular matrix (ECM). METHODS: B6 mice received daily intraperitoneal (IP) injections of VIP, while control mice were similarly injected with sterile phosphate buffered saline (PBS). Real-time RT-PCR, ELISA, and immunofluorescent staining were used to assess the effects of VIP treatment on ECM molecule expression after Pseudomonas aeruginosa-induced keratitis. We also compared the effect of VIP treatment on lipopolysaccharide (LPS)-stimulated B6- and BALB/c-derived fibroblasts. RESULTS: In vivo analyses revealed that VIP treatment of P. aeruginosa-infected B6 corneas led to a significant increase in ECM molecules associated with healing/homeostasis, while those associated with ECM degradation were significantly down-regulated when compared to wild-type (WT) controls. In vitro studies revealed that VIP treatment of lipopolysaccharide-stimulated fibroblasts derived from susceptible B6 and resistant BALB/c mice expressed distinct differences in ECM molecule expression, whereby the latter expressed higher levels of ECM molecules aimed at reconstitution. Furthermore, differential expression of VIP receptor-1/VIP receptor-2 (VIPR1/VIPR2) was observed between B6 and BALB/c after VIP treatment of LPS-stimulated fibroblasts. CONCLUSIONS: VIP treatment functions to enhance ECM reconstitution, which appears to be carried out in large part by fibroblasts via VIPR2. Overall, the data from this study suggest that VIP not only regulates disease pathogenesis, but also functions to restore integrity of the corneal stroma.


Assuntos
Córnea/patologia , Infecções Oculares Bacterianas/tratamento farmacológico , Ceratite/tratamento farmacológico , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/isolamento & purificação , Peptídeo Intestinal Vasoativo/farmacologia , Animais , Córnea/efeitos dos fármacos , Córnea/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Infecções Oculares Bacterianas/metabolismo , Infecções Oculares Bacterianas/microbiologia , Feminino , Homeostase , Ceratite/metabolismo , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , RNA Mensageiro/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/genética , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Mol Pharmacol ; 77(1): 95-101, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19854890

RESUMO

The neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) and their class II G protein-coupled receptors VPAC(1), VPAC(2), and PAC(1) play important roles in human physiology. No small molecule modulator has ever been reported for the VIP/PACAP receptors, and there is a lack of specific VPAC(2) antagonists. Via high-throughput screening of 1.67 million compounds, we discovered a single small molecule antagonist of human VPAC(2), compound 1. Compound 1 inhibits VPAC(2)-mediated cAMP accumulation with an IC(50) of 3.8 microM and the ligand-activated beta-arrestin2 binding with an IC(50) of 2.3 microM. Compound 1 acts noncompetitively in Schild analysis. It is a specific VPAC(2) antagonist with no detectable agonist or antagonist activities on VPAC(1) or PAC(1). Compound 2, a close structural analog of compound 1, was also found to be weakly active. To our surprise, compound 1 is completely inactive on the closely related mouse VPAC(2). Chimera experiments indicate that compounds 1 and 2 bind to the seven transmembrane (7TM) region of the receptor as opposed to the N-terminal extracellular domain, where the natural ligand binds. Compound 1, being the first small molecular antagonist that is specific for VPAC(2), and the only VPAC(2) antagonist molecule known to date that allosterically interacts with the 7TM region, will be a valuable tool in further study of VPAC(2) and related receptors. This study also highlights the opportunities and challenges facing small molecule drug discovery for class II peptide G protein-coupled receptors.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Animais , Sítios de Ligação , AMP Cíclico/metabolismo , Humanos , Concentração Inibidora 50 , Camundongos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos
9.
Neuroreport ; 20(10): 957-62, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19474768

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) has neuroprotective properties and plays an important role in neuroinflammation. PACAP38 interacts with its receptors, PAC1, and VPAC, on astrocytes at 10(-8) M to induce biphasic Ca2+ transients, which were reduced to a single transient by the PAC1-blocking PACAP antagonist PACAP6-38. At 10(-12) M even the single transient, corresponding to PAC1 was blocked. PACAP-induced Ca2+ transients were more pronounced in astrocytes cocultured with brain endothelial cells than in monocultured astrocytes, indicating that astrocytes that receive signals from microvessels develop more sensitive signal transduction systems for Ca. In this sensitive system, PACAP38 attenuated 5-HT, histamine, and ATP-evoked Ca2+ transients, showing the anti-inflammatory properties of PACAP.


Assuntos
Astrócitos/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Encefalite/tratamento farmacológico , Gliose/tratamento farmacológico , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Sinalização do Cálcio/fisiologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Encefalite/metabolismo , Encefalite/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Gliose/metabolismo , Gliose/fisiopatologia , Histamina/metabolismo , Histamina/farmacologia , Microcirculação/efeitos dos fármacos , Microcirculação/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Serotonina/metabolismo , Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Neuropharmacology ; 51(6): 1086-98, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16930633

RESUMO

VPAC/PAC receptor activation classically results in cyclic-AMP production, with limited reports evaluating calcium signalling. These studies systematically characterise intracellular cyclic-AMP ([cAMP](i)) and calcium ([Ca(2+)](i)) responses in CHO-cells expressing recombinant human (h) VPAC/PAC receptors (hVPAC(1)R, hVPAC(2)R, hPAC(1)R), using two simple, non-radioactive, HT-amenable assays. The rank order of potency (ROP) of the agonists VIP, PACAP-27 and PACAP-38 was similar in both assays for each individual receptor subtype, although potencies (EC(50)) in the [Ca(2+)](i) assay were approximately 100-fold lower. Importantly, this shift was also evident in SHSY-5Y cells endogenously expressing hPAC(1)R. Furthermore, [Ala(11,22,28)]VIP and maxadilan were selective hVPAC(1)R and hPAC(1)R agonists, respectively, and although R3P65 had no demonstrable hVPAC(2)R selectivity, these compounds exhibited comparable reductions in [Ca(2+)](i) EC(50) values. In contrast, PG97-269 and PG99-465, putatively selective hVPAC(1)R and hVPAC(2)R antagonists, respectively, were marginally less potent in [cAMP](i) studies, whereas M65 was equipotent at hPAC(1)R. Moreover, PG99-465 alone increased [cAMP](i) at all three hVPAC/PAC receptor subtypes, with full hVPAC(1)R and hPAC(1)R agonism. With equivalent agonist ROPs generated in both assays, [Ca(2+)](i) signalling provides an alternative approach to examine hVPAC/PAC receptor pharmacology. However, these studies underscore the paucity of receptor selective compounds, complexities in comparing drug potencies across assays, and the pleiotropic nature of VPAC/PAC-receptor signalling.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , AMP Cíclico/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Interpretação Estatística de Dados , Ensaio de Imunoadsorção Enzimática , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/agonistas , Receptores Tipo II de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/agonistas , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/antagonistas & inibidores , Transfecção , Peptídeo Intestinal Vasoativo/análogos & derivados , Peptídeo Intestinal Vasoativo/farmacologia
11.
J Cereb Blood Flow Metab ; 26(1): 58-67, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15959462

RESUMO

Vasoactive intestinal peptide (VIP) is a vasodilator peptide present in cerebrovascular nerves. Vasoactive intestinal peptide can activate VPAC1, VPAC2 and the NPR-C receptor. This study sought to determine the receptors involved in VIP-induced vasodilation of porcine basilar arteries. Porcine basilar arteries contained the messenger ribonucleic acid of all three receptors. Immunocytochemical analysis of porcine basilar arteries revealed that the VPAC1 receptor is expressed on the endothelium, VPAC2 on the outer layers of the media and the NPR-C receptor throughout the artery, including nerves. Vasodilator responses to all receptor agonists showed that the receptors are functional. The vasodilator response to the VPAC1 receptor agonist was inhibited by L-NAME and abolished by endothelial denudation. Vasodilation induced by Ro-25-1553, the VPAC2 agonist, was unaffected by NOS inhibition or removal of the endothelium. Activation of the NPR-C receptor produced a vasodilation, which was susceptible to NOS inhibition and independent of endothelium. The vasodilator response to electrical stimulation at 20 Hz was attenuated by PG-99-465, the VPAC2 antagonist. This study shows that all known VIP receptors are involved in VIP-mediated vasodilation of porcine basilar arteries. The VPAC1 receptor is located on the endothelium and elicits vasodilation by generating nitric oxide (NO). The VPAC2 receptor is mainly expressed in the outer layers of the smooth muscle and induces vasodilation independently of NO in response to VIP released from intramural nerves. The NPR-C receptor produces NO-dependent vasodilation independently of the endothelium by stimulation of nNOS in intramural nerves.


Assuntos
Artéria Basilar/efeitos dos fármacos , Receptores do Fator Natriurético Atrial/fisiologia , Receptores Tipo II de Peptídeo Intestinal Vasoativo/fisiologia , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/fisiologia , Peptídeo Intestinal Vasoativo/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Artéria Basilar/fisiologia , Imuno-Histoquímica , Receptores do Fator Natriurético Atrial/efeitos dos fármacos , Receptores Tipo II de Peptídeo Intestinal Vasoativo/biossíntese , Receptores Tipo II de Peptídeo Intestinal Vasoativo/efeitos dos fármacos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/biossíntese , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA