Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 12: 711906, 2021.
Article in English | MEDLINE | ID: mdl-34867774

ABSTRACT

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.


Subject(s)
Allosteric Regulation/drug effects , Drug Repositioning/methods , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Ticagrelor/pharmacology , Binding Sites , Computer Simulation , Drug Evaluation, Preclinical/methods , Molecular Structure , Protein Conformation/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/chemistry , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/chemistry , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Ticagrelor/chemistry
2.
Neurobiol Learn Mem ; 180: 107423, 2021 04.
Article in English | MEDLINE | ID: mdl-33705861

ABSTRACT

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.


Subject(s)
Basolateral Nuclear Complex/drug effects , CA1 Region, Hippocampal/drug effects , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Recognition, Psychology/drug effects , Social Perception/drug effects , Animals , Basolateral Nuclear Complex/metabolism , CA1 Region, Hippocampal/metabolism , Memory Consolidation/drug effects , Nitric Oxide Donors/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Recognition, Psychology/physiology , S-Nitroso-N-Acetylpenicillamine/pharmacology
3.
J Leukoc Biol ; 100(6): 1385-1393, 2016 12.
Article in English | MEDLINE | ID: mdl-27381006

ABSTRACT

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.


Subject(s)
Macrophages/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/physiology , Receptors, Vasoactive Intestinal Polypeptide, Type I/physiology , Vasoactive Intestinal Peptide/pharmacology , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cyclic AMP/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Inflammation , Macrophage Activation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/metabolism , RNA, Messenger/biosynthesis , Receptors, Vasoactive Intestinal Peptide, Type II/biosynthesis , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/biosynthesis , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Synovial Fluid/cytology , Up-Regulation
4.
J Neurosci ; 35(50): 16463-78, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674871

ABSTRACT

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.


Subject(s)
Dopaminergic Neurons/drug effects , Dopaminergic Neurons/immunology , MPTP Poisoning/drug therapy , MPTP Poisoning/immunology , Neuroprotective Agents/therapeutic use , Oligopeptides/pharmacology , Receptors, Vasoactive Intestinal Peptide/agonists , Animals , CD4-Positive T-Lymphocytes/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cytokines/metabolism , Humans , Immunohistochemistry , MPTP Poisoning/physiopathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Oligopeptides/pharmacokinetics , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Spleen/cytology , Spleen/drug effects , Substantia Nigra/enzymology , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
5.
Invest Ophthalmol Vis Sci ; 53(12): 7432-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23036997

ABSTRACT

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.


Subject(s)
Cornea/pathology , Eye Infections, Bacterial/drug therapy , Keratitis/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/isolation & purification , Vasoactive Intestinal Peptide/pharmacology , Animals , Cornea/drug effects , Cornea/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Eye Infections, Bacterial/metabolism , Eye Infections, Bacterial/microbiology , Female , Homeostasis , Keratitis/metabolism , Keratitis/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , RNA, Messenger/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Am J Physiol Cell Physiol ; 294(2): C477-87, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077607

ABSTRACT

The smooth muscle of the gut expresses mainly G(s) protein-coupled vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating peptide receptors (VPAC(2) receptors), which belong to the secretin family of G protein-coupled receptors. The extent to which PKA and G protein-coupled receptor kinases (GRKs) participate in homologous desensitization varies greatly among the secretin family of receptors. The present study identified the novel role of PKA in homologous desensitization of VPAC(2) receptors via the phosphorylation of GRK2 at Ser(685). VIP induced phosphorylation of GRK2 in a concentration-dependent fashion, and the phosphorylation was abolished by blockade of PKA with cell-permeable myristoylated protein kinase inhibitor (PKI) or in cells expressing PKA phosphorylation-site deficient GRK2(S685A). Phosphorylation of GRK2 increased its activity and binding to G betagamma. VIP-induced phosphorylation of VPAC(2) receptors was abolished in muscle cells expressing kinase-deficient GRK2(K220R) and attenuated in cells expressing GRK2(S685A) or by PKI. VPAC(2) receptor internalization (determined from residual (125)I-labeled VIP binding and receptor biotinylation after a 30-min exposure to VIP) was blocked in cells expressing GRK2(K220R) and attenuated in cells expressing GRK2(S685A) or by PKI. Finally, VPAC(2) receptor degradation (determined from residual (125)I-labeled VIP binding and receptor expression after a prolonged exposure to VIP) and functional VPAC(2) receptor desensitization (determined from the decrease in adenylyl cyclase activity and cAMP formation after a 30-min exposure to VIP) were abolished in cells expressing GRK2(K220R) and attenuated in cells expressing GRK2(S685A). These results demonstrate that in gastric smooth muscle VPAC(2) receptor phosphorylation is mediated by GRK2. Phosphorylation of GRK2 by PKA enhances GRK2 activity and its ability to induce VPAC(2) receptor phosphorylation, internalization, desensitization, and degradation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Endocytosis/physiology , G-Protein-Coupled Receptor Kinase 2/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Animals , Binding, Competitive/physiology , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Endocytosis/drug effects , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , G-Protein-Coupled Receptor Kinase 2/chemistry , G-Protein-Coupled Receptor Kinase 2/genetics , Gastric Mucosa/metabolism , Mutation/genetics , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Rabbits , Radioligand Assay , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Stomach/cytology , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
7.
Neuropharmacology ; 51(6): 1086-98, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16930633

ABSTRACT

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.


Subject(s)
Calcium Signaling/drug effects , Cyclic AMP/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Amino Acid Sequence , Animals , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetinae , Data Interpretation, Statistical , Enzyme-Linked Immunosorbent Assay , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Vasoactive Intestinal Peptide, Type II/agonists , Receptors, Vasoactive Intestinal Peptide, Type II/antagonists & inhibitors , Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists , Receptors, Vasoactive Intestinal Polypeptide, Type I/antagonists & inhibitors , Transfection , Vasoactive Intestinal Peptide/analogs & derivatives , Vasoactive Intestinal Peptide/pharmacology
8.
J Neurophysiol ; 96(2): 858-71, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16641377

ABSTRACT

Thalamic nuclei can generate intrathalamic rhythms similar to those observed at various arousal levels and pathophysiological conditions such as absence epilepsy. These rhythmic activities can be altered by a variety of neuromodulators that arise from brain stem regions as well as those that are intrinsic to the thalamic circuitry. Vasoactive intestinal peptide (VIP) is a neuropeptide localized within the thalamus and strongly attenuates intrathalamic rhythms via an unidentified receptor subtype. We have used transgenic mice lacking a specific VIP receptor, VPAC(2), to identify its role in VIP-mediated actions in the thalamus. VIP strongly attenuated both the slow, 2-4 Hz and spindle-like 5-8 Hz rhythmic activities in slices from wild-type mice (VPAC(2)(+/+)) but not in slices from VPAC(2) receptor knock-out mice (VPAC(2)(-/-)), which suggests a major role of VPAC(2) receptors in the antioscillatory actions of VIP. Intracellular recordings revealed that VIP depolarized all relay neurons tested from VPAC(2)(+/+) mice. In VPAC(2)(-/-) mice, however, VIP produced no membrane depolarization in 80% of neurons tested. In relay neurons from VPAC(2)+/+ mice, VIP enhanced the hyperpolarization-activated mixed cation current, I(h), via cyclic AMP activity, but VIP did not alter I(h) in VPAC(2)-/- mice. In VPAC(2)-/- mice, pituitary adenylate cyclase activating-polypeptide (PACAP) depolarized the majority of relay neurons via I(h) enhancement presumably via PAC(1) receptor activation. Our findings suggest that VIP-mediated actions are predominantly mediated by VPAC(2) receptors, but PAC(1) receptors may play a minor role. The excitatory actions of VIP and PACAP suggest these peptides may not only regulate intrathalamic rhythmic activities, but also may influence information transfer through thalamocortical circuits.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Interneurons/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Thalamus/cytology , Thalamus/drug effects , Vasoactive Intestinal Peptide/pharmacology , Animals , Cyclic AMP/physiology , Electrophysiology , Extracellular Space/drug effects , Extracellular Space/physiology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Second Messenger Systems/physiology
9.
J Cereb Blood Flow Metab ; 26(1): 58-67, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15959462

ABSTRACT

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.


Subject(s)
Basilar Artery/drug effects , Receptors, Atrial Natriuretic Factor/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/physiology , Receptors, Vasoactive Intestinal Polypeptide, Type I/physiology , Vasoactive Intestinal Peptide/pharmacology , Vasodilation/drug effects , Animals , Basilar Artery/physiology , Immunohistochemistry , Receptors, Atrial Natriuretic Factor/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/biosynthesis , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Receptors, Vasoactive Intestinal Polypeptide, Type I/biosynthesis , Receptors, Vasoactive Intestinal Polypeptide, Type I/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Swine
10.
Mol Cell Neurosci ; 30(4): 523-37, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16202621

ABSTRACT

Activation of intracellular signaling pathways involving p38 and p42/44 MAP kinases may contribute importantly to synaptic plasticity underlying spinal neuronal sensitization. Inhibitors of p38 or p42/44 pathways moderately attenuated responses of dorsal horn neurons evoked by mustard oil but not brush and alleviated the behavioral reflex sensitization seen following nerve injury. Activation of p38 and p42/44 MAP kinases in spinal cord ipsilateral to constriction injury was reduced by antagonists of NMDA, VPAC2 and NK2 (but not related) receptors, the glial inhibitor propentofylline and inhibitors of TNF-alpha. A VPAC2 receptor agonist enhanced p38 phosphorylation and caused behavioral reflex sensitization in naïve animals that could be blocked by co-administration of p38 inhibitor. Conversely, an NK2 receptor agonist activated p42/44 and caused behavioral sensitization that could be prevented by co-administration of p42/44 inhibitor. Thus, spinal p38 and p42/44 MAP kinases are activated in neuropathic pain states by mechanisms involving VPAC2, NK2, NMDA receptors and glial cytokine production.


Subject(s)
MAP Kinase Signaling System/physiology , Neuroglia/metabolism , Peripheral Nervous System Diseases/metabolism , Posterior Horn Cells/metabolism , Receptors, Neurokinin-2/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Animals , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Inflammation Mediators/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Neuralgia/metabolism , Neuralgia/physiopathology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Peripheral Nerve Injuries , Peripheral Nerves/metabolism , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Phosphorylation/drug effects , Physical Stimulation , Posterior Horn Cells/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neurokinin-2/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Xanthines/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...