pacap receptors
Recently Published Documents


TOTAL DOCUMENTS

65
(FIVE YEARS 6)

H-INDEX

21
(FIVE YEARS 1)

2021 ◽  
Vol 12 ◽  
Author(s):  
Ingrid Langer ◽  
Dorota Latek

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.


2021 ◽  
Vol 11 ◽  
Author(s):  
Tibor Hajdú ◽  
Patrik Kovács ◽  
Emese Zsigrai ◽  
Roland Takács ◽  
Judit Vágó ◽  
...  

Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide which is distributed throughout the body. PACAP influences development of various tissues and exerts protective function during cellular stress and in some tumour formation. No evidence is available on its role in neural crest derived melanocytes and its malignant transformation into melanoma. Expression of PACAP receptors was examined in human skin samples, melanoma lesions and in a primary melanocyte cell culture. A2058 and WM35 melanoma cell lines, representing two different stages of melanoma progression, were used to investigate the effects of PACAP. PAC1 receptor was identified in melanocytes in vivo and in vitro and in melanoma cell lines as well as in melanoma lesions. PACAP administration did not alter viability but decreased proliferation of melanoma cells. With live imaging random motility, average speed, vectorial distance and maximum distance of migration of cells were reduced upon PACAP treatment. PACAP administration did not alter viability but decreased proliferation capacity of melanoma cells. On the other hand, PACAP administration decreased the migration of melanoma cell lines towards fibronectin chemoattractant in the Boyden chamber. Furthermore, the presence of the neuropeptide inhibited the invasion capability of melanoma cell lines in Matrigel chambers. In summary, we provide evidence that PACAP receptors are expressed in melanocytes and in melanoma cells. Our results also prove that various aspects of the cellular motility were inhibited by this neuropeptide. On the basis of these results, we propose PACAP signalling as a possible target in melanoma progression.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Parleen K Pandher ◽  
Ekaterina Filatov ◽  
Sarah L Gray

Abstract Pituitary adenylate cyclase-activating polypeptide (PACAP) is being studied to understand the endocrine regulation of energy balance and has been shown to be important in the regulation of the stress response (1,2). Specifically, PACAP has been shown to regulate thermogenesis, an energy burning process regulated by the sympathetic nervous system that contributes to achieving energy homeostasis in response to cold stress and overfeeding. PACAP is expressed in the sympathetic nervous system and is required at the adrenomedullary synapse to maintain epinephrine secretion from the adrenal medulla in response to physiological stress (3). Across the branches of the sympathetic nervous system, PACAP receptor expression is most well characterized in the superior cervical ganglia (SCG) (4). However, a detailed characterization of PACAP and its receptors has not been performed in ganglia whose postganglionic fibres innervate adipose tissues (stellate and celiac ganglia) in response to thermogenic stress. We hypothesized that PACAP is produced by preganglionic neurons innervating the stellate and celiac ganglia, and act on PACAP receptors expressed on the post-ganglionic neurons, and this expression will be upregulated in response to chronic cold stress. Due to their small and amorphous shape, we have developed a protocol to reliably isolate the stellate and celiac ganglia and validate their identity through the presence of tyrosine hydroxylase mRNA, using adrenal and SCG samples as positive controls. PACAP receptor expression (VPAC1, VPAC2, PAC1) was examined in the ganglia utilizing real-time PCR, and PACAP protein was visualized in the ganglia of transgenic mice that express eGFP under the control of the PACAP promoter (PACAP-eGFP mice) (5). This research demonstrates the expression of PACAP receptors in ganglia whose postganglionic fibres innervate adipose tissue, enhancing our understanding of PACAP’s role in the SNS, and its contribution to the regulation of adaptive thermogenesis. References: (1) Gray et al., Pacap: Regulator of the stress response. In: Fink G, ed. Stress: Physiology, biochemistry, and pathology. 2019:279-291. (2) Mustafa, Adv Pharmacol. San Diego, Calif:445-457. (3) Eiden et al., Pflungers Arch. 2018 Jan;470(1):79-88. (4) Braas et al., J Biol Chem. 1999 Sep 24;274(39):27702-27710. (5) Condro et al., J Comp Neurol. 2016 Dec 15; 524(18):3827-3848.


2019 ◽  
Vol 2019 (4) ◽  
Author(s):  
Jan Fahrenkrug ◽  
Edward J. Goetzl ◽  
Illana Gozes ◽  
Anthony Harmar ◽  
Marc Laburthe ◽  
...  

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) receptors (nomenclature as agreed by the NC-IUPHAR Subcommittee on Vasoactive Intestinal Peptide Receptors [64, 65]) are activated by the endogenous peptides VIP, PACAP-38, PACAP-27, peptide histidine isoleucineamide (PHI), peptide histidine methionineamide (PHM) and peptide histidine valine (PHV). VPAC1 and VPAC2 receptors display comparable affinity for the PACAP peptides, PACAP-27 and PACAP-38, and VIP, whereas PACAP-27 and PACAP-38 are >100 fold more potent than VIP as agonists of most isoforms of the PAC1 receptor. However, one splice variant of the human PAC1 receptor has been reported to respond to PACAP-38, PACAP-27 and VIP with comparable affinity [29]. PG 99-465 [115] has been used as a selective VPAC2 receptor antagonist in a number of physiological studies, but has been reported to have significant activity at VPAC1 and PAC1 receptors [35]. The selective PAC1 receptor agonist maxadilan, was extracted from the salivary glands of sand flies (Lutzomyia longipalpis) and has no sequence homology to VIP or the PACAP peptides [116]. Two deletion variants of maxadilan, M65 [180] and Max.d.4 [117] have been reported to be PAC1 receptor antagonists, but these peptides have not been extensively characterised.


2019 ◽  
Vol 110 (3-4) ◽  
pp. 271-281
Author(s):  
Matthew M. Hurley ◽  
Eden M. Anderson ◽  
Christopher Chen ◽  
Brian Maunze ◽  
Evan M. Hess ◽  
...  

Leptin signaling pathways, stemming primarily from the hypothalamus, are necessary for maintaining normal energy homeostasis and body weight. In both rodents and humans, dysregulation of leptin signaling leads to morbid obesity and diabetes. Since leptin resistance is considered a primary factor underlying obesity, understanding the regulation of leptin signaling could lead to therapeutic tools and provide insights into the causality of obesity. While leptin actions in some hypothalamic regions such as the arcuate nuclei have been characterized, less is known about leptin activity in the hypothalamic ventromedial nuclei (VMN). Recently, pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to reduce feeding behavior and alter metabolism when administered into the VMN in a pattern similar to that of leptin. In the current study, we examined whether leptin and PACAP actions in the VMN share overlapping pathways in the regulation of energy balance. Interestingly, PACAP administration into the VMN increased STAT3 phosphorylation and SOCS3 mRNA expression, both of which are hallmarks of leptin receptor activation. In addition, BDNF mRNA expression in the VMN was increased by both leptin and PACAP administration. Moreover, antagonizing PACAP receptors fully reversed the behavioral and cellular effects of leptin injections into the VMN. Electrophysiological studies further illustrated that leptin-induced effects on VMN neurons were blocked by antagonizing PACAP receptors. We conclude that leptin dependency on PACAP signaling in the VMN suggests a potential common signaling cascade, allowing a tonically and systemically secreted neuropeptide to be more precisely regulated by central neuropeptides.


2016 ◽  
Vol 299 (9) ◽  
pp. 1293-1299 ◽  
Author(s):  
Yuko Matoba ◽  
Naoko Nonaka ◽  
Yoshitoki Takagi ◽  
Eisaku Imamura ◽  
Masayuki Narukawa ◽  
...  

2015 ◽  
Vol 47 (2) ◽  
pp. 923-932 ◽  
Author(s):  
Bartolomeo Gorgoglione ◽  
Yamila Carpio ◽  
Christopher J. Secombes ◽  
Nick G.H. Taylor ◽  
Juana María Lugo ◽  
...  

2015 ◽  
Vol 2 (Suppl 1) ◽  
pp. A19
Author(s):  
MJ Müller ◽  
A Kislat ◽  
A Hippe ◽  
A Poppe ◽  
S Goldmann ◽  
...  

Neuropeptides ◽  
2013 ◽  
Vol 47 (2) ◽  
pp. 85-92 ◽  
Author(s):  
André Erdling ◽  
Majid Sheykhzade ◽  
Aida Maddahi ◽  
Ferenc Bari ◽  
Lars Edvinsson

Sign in / Sign up

Export Citation Format

Share Document