VIP and PACAP receptors: Introduction

General

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are members of a superfamily of structurally related peptide hormones that includes glucagon, glucagon-like peptides, secretin, gastric inhibitory peptide (GIP) and growth hormone-releasing hormone (GHRH). At least three receptors for PACAP exist in mammals, two of which are also high-affinity receptors for VIP [40].

VIP

VIP, first isolated from porcine intestine as a 28-amino acid (aa) peptide capable of inducing vasodilatation in the canine femoral artery [106-107], has subsequently been shown to have many other actions as a neuroendocrine hormone, putative neurotransmitter and cytokine. The presence of VIP and specific VIP binding sites in defined pathways in the brain indicate that it may play an important role in central nervous system (CNS) function [10,70]. VIP is now widely accepted as a co-transmitter, with nitric oxide and carbon monoxide, of nonadrenergic, noncholinergic relaxation of both vascular and nonvascular smooth muscle [108]. VIP may also promote neuronal survival [11] and regulate glycogen metabolism in the cerebral cortex [113]. VIP stimulates prolactin secretion from the pituitary [98] and catecholamine release from the adrenal medulla [69]. In the immune system, VIP regulates T cell traffic and inhibits mitogen-activated proliferation of T cells by inhibiting interleukin-2 production [84]. Other actions of VIP include stimulation of electrolyte secretion and protection against oxidant injury [31,104-105].

In common with the precursors of a number of other neuroendocrine peptides, the VIP precursor polypeptide (prepro-VIP) contains sequences encoding several additional biologically active peptides, including either peptide histidine isoleucine (PHI: in non-human mammals) [121] or peptide histidine methionine (PHM: the human equivalent of PHI) [49] and peptide histidine valine (PHV), a C-terminally extended form of PHI and PHM [146].

PACAP

PACAP was first identified as a 38aa peptide (PACAP-38) from ovine hypothalamus that stimulated adenylate cyclase in rat anterior pituitary cells in culture [73]. Subsequently, a C-terminally truncated, 27aa form of the peptide (PACAP-27) was isolated from the same source [74]. In the CNS, PACAP and the mRNA encoding its precursor are most abundant in the hypothalamus, with lower levels in many other brain regions [23]. PACAP is also present in a number of peripheral tissues, including the gastrointestinal tract, adrenal gland and testis [4,23]. Although first isolated as a hypophysiotropic hormone, the role of PACAP in the regulation of pituitary hormone secretion is still poorly understood [96]. However, in the CNS, PACAP released from retinal afferents to the rat suprachiasmatic nucleus has been proposed to function as a daytime regulator of the biological clock [37]. PACAP is expressed in sympathetic neurons and in the cholinergic innervation of the adrenal medulla, where it is though to facilitate prolonged secretion of catecholamines under conditions of high stress [35,94]. PACAP is also thought to regulate exocrine and endocrine secretion from the pancreas [144]. For a recent review of the structure and functions of PACAP and its receptors, see [133].

Receptor types

Ligand binding studies [112] suggested the existence of at least two distinct receptors for PACAP, one with much greater affinity for PACAP than for VIP (the 'PACAP type I receptor') and a second with high affinity for both PACAP and VIP (the 'PACAP type II receptor'). Based on the relative potencies of natural and synthetic VIP analogues, it was later suggested that two types of high affinity VIP (PACAP type II) receptors existed in rat and human tissues. In addition to the 'classical' VIP receptors from intestinal cells [60], a second receptor was identified in the human SUP-T1 lymphoblast cell line [102] and in lung cancer cell lines [66]. Subsequently, two high-affinity receptors for both VIP and PACAP ('PACAP type II receptors', now referred to as VPAC1 and VPAC2) and a third receptor selective for PACAP (the 'PACAP type I receptor', now referred to as PAC1) were cloned. Progress in characterising the functions of the three receptor types has been hindered by the limited number of selective pharmacological tools available. Briefly, [Ala11,22,28]VIP and [K15, R16, L17]VIP(3-7)/GRF(8-27) are selective agonists of the VPAC1 receptor and PG 97-269 is a selective antagonist. Ro 25-1392 is a VPAC2 agonist but there is no highly selective VPAC2 antagonist yet. Maxadilan is an agonist of PAC1 and Max.d.4 (maxadilan Δ24-42) and M65 (maxadilan Δ25-41) are PAC1 antagonists but the use of these peptides has been limited due to problems with their availability. Finally, it is important to note that although PACAP(6-38) has been used as a PAC1 receptor antagonist in many studies, it does not discriminate between PAC1 and VPAC2 receptors.

VPAC receptors

For recent critical reviews of the pharmacology and signalling properties of VPAC1 and VPAC2 receptors, see [61-63].

The VPAC1 receptor, which responds to VIP and PACAP with comparable affinity ('PACAP type II' pharmacology) was first isolated from rat lung [48]. This receptor is widely distributed in the CNS, most abundantly in the cerebral cortex and hippocampus [48,126], in peripheral tissues including liver, lung and intestine [42,46,48,55,100-101,115,126] and in T lymphocytes [20]. There are important differences between species in the pharmacology of VPAC1 receptors [16]. Selective VPAC1 receptor agonists [28,80] and antagonists [27] have been described.

The VPAC2 receptor, first cloned from rat olfactory bulb [67] also responds to VIP and PACAP with comparable affinity ('PACAP type II' pharmacology). In the CNS, the highest concentrations of messenger RNA encoding the VPAC2 receptor are found in the thalamus and suprachiasmatic nucleus (SCN) and lower levels in the hippocampus, brainstem, spinal cord and dorsal root ganglia [110,126]. The receptor is also present in many peripheral tissues, including smooth muscles in the cardiovascular, gastrointestinal and reproductive systems [3,42,47,58,100-101,126,142]. A number of selective peptide agonists [29,82-83,125,143] and an antagonist with some selectivity for the VPAC2 receptor [76] have been described.

The tissue distribution of VPAC1 and VPAC2 receptors can be determined by in vitro receptor autoradiography using [125I]-VIP as the radioligand and displacement with the VPAC1 selective agonist [K15,R16,L17]VIP(3-7)/GRF(8-27) (KRL-VIP) and the VPAC2 selective agonist Ro25-1553 to distinguish the two receptor types [42,100-101]. [125I]-Ro25-1553 can also be used to localise VPAC2 receptors [42,100-101,136].

PAC receptors

The PAC1 receptor, first cloned from a rat pancreatic acinar carcinoma cell line [91] recognises PACAP-27 and PACAP-38 with much higher potency than VIP. Messenger RNA encoding this receptor is expressed predominantly in the CNS (most abundantly in the olfactory bulb, thalamus, hypothalamus, the dentate gyrus of the hippocampus and in granule cells of the cerebellum [43,111]). The receptor is also expressed abundantly in the embryonic nervous system [109,141,148] and in a number of peripheral tissues, most abundantly in the adrenal medulla [75,100-101,112,114]. There is apparent heterogeneity of PAC1 receptors in tissues and cell lines, where two types of 'PACAP type I' pharmacology have been observed: type IA receptors, with high affinity for both PACAP-27 and PACAP-38; and type IB receptors, with high affinity for PACAP-38 but low affinity for PACAP-27 [103,112]. The difference between the two receptor subtypes may reflect differences in G protein-coupling and second messenger mechanisms [128] or result from alternative splicing of PAC1 receptor mRNA [88,114].

Unlike the VPAC1 and VPAC2 receptors, the PAC1 receptor has numerous splice variants. Within the part of the PAC1 receptor cDNA encoding the third intracellular loop, splice variants either containing or lacking each of two alternative exons ('hip' and 'hop') exist. The hop exon exists in two forms (hop1 and hop2) as the result of the existence of two alternative splice acceptor sites three nucleotides apart. Thus, six possible splice variants, which differ in their intracellular signal transduction pathways can be generated [53,114]. Four variants of the human PAC1 receptor (null, SV-1, SV-2 and SV-3) resulting from alternative splicing of sequences equivalent to hip and hop1 have also been described [92] and were shown to differ in their ability to activate phospholipase C (PLC). In addition, splice variation in the N-terminal extracellular domain of the PAC1 receptor has been reported. Splicing out of the 4th and 5th coding exons, leading to a 21aa deletion, has been reported in human and mouse [19,88]. Surprisingly, the human splice variant bound PACAP-27, PACAP-38 and VIP with similar high affinity and all three peptides stimulated cyclic AMP accumulation with similar potency [19]. Additional N-terminal splice variants resulting from splicing out of the 3rd, 4th and 5th exons of the human gene [19] and by insertion of an additional 72 base pairs encoding 24aa (exon 3a) between coding exons 3 and 4 [18] have also been described.

Receptor structure and activation

PAC1, VPAC1 and VPAC2 receptors belong to the family B, also referred to as class II, of GPCRs. This family comprises receptors for all peptides structurally related to VIP and PACAP, and also receptors for parathyroid hormone, corticotropin-releasing factor, calcitonin and related peptides [15,39]. For all family B receptors, the large N-terminal ectodomain plays a crucial role in ligand recognition, prompting structural studies of this domain [15,61,63]. As initially described for the mouse CRF2 receptor [32], the structure comprises a crucial Sushi domain characterized by two antiparallel β sheets and stabilized by three disulphide bonds and a salt bridge sandwiched between aromatic rings of two tryptophan residues. Structures of the ectodomains of PAC1 receptor [59,117] and VPAC2 receptors (PDB ID: 2X57) have been determined by x-ray or NMR and a structural model of the VPAC1 receptor obtained by homology modelling associated with photoaffinity experiments [120]. The data are consistent with the two-site model for peptide binding to family B GPCRs in which the C-terminal and central α-helical parts of the peptide hormone interact with the Sushi domain in the N-terminal ectodomain (N-ted) ultimately positioning the N-terminus of the peptide to contact the transmembrane region resulting in receptor activation [15,61,63]. This latter contact region remains elusive since none structure for full-length family B GPCRs has been determined yet. However, the presence of a helix N-capping motif in cognate peptide ligands of all family B receptors, including VIP and PACAP, supports that the folded backbone conformation of a N-cap is formed upon receptor binding and constitutes a key element underlying family B GPCR activation [79].

Functions

The widespread distribution of VIP and PACAP and their receptors in the brain and periphery has led to many hypotheses concerning the physiological functions of these receptors. However, the availability of mutant mice lacking VIP [13], PACAP [14,56], the VPAC2 receptor [5,41], the VPAC1 receptor [21] and the PAC1 receptor [38,52,87,118] has permitted experimental validation of a number of physiological functions for these receptors.

Both VIP and PACAP play roles in the control of circadian rhythms in the brain's "master clock" in the suprachiasmatic nuclei (SCN) of the hypothalamus. Light entrains the SCN clock through a population of retinal ganglion cells that project to the SCN via the retinohypothalamic tract and contain both glutamate and PACAP. Studies of knockout mice lacking the PAC1 receptor or its ligand PACAP [14,38,56] show that PACAP plays a role in modulating the light-induced resetting of the behavioural rhythm and light-induced clock gene expression in the SCN. In contrast, VIP is synthesised in a population of SCN neurones, many of which are thought to receive a direct retinal innervation, and acts on VPAC2 receptors, which are expressed throughout the SCN. Studies of knockout mice lacking the VPAC2 receptor indicate that this receptor is necessary for the generation of normal circadian rhythms of electrical activity, clock gene expression and behaviour [6,17,41,45]. VIP deficient mice also display a severely disrupted circadian phenotype, sharing many common features with that of VPAC2 receptor null mice [6,13].

Both VIP and PACAP, acting through PAC1 and VPAC2 receptors on pancreatic β-cells, have been implicated in the control of pancreatic insulin secretion. PAC1 receptor-deficient mice display impaired insulinotropic response to glucose, reduced glucose tolerance and impaired glucagon response to insulin-induced hypoglycaemia [50,89] and overexpression of overexpression of PACAP in mouse pancreatic β-cells has been reported to enhance insulin secretion and ameliorate streptozotocin-induced diabetes [145] and to inhibit hyperinsulinemia and islet hyperplasia in agouti yellow mice [122]. VPAC2 receptor null mice have been reported to be able to maintain a normal response to glucose challenge with lower levels of insulin than wild type mice, suggesting a significant increase in insulin sensitivity in the knockout mice [5]. A selective peptide agonist of the VPAC2 receptor stimulated glucose-dependent insulin secretion in isolated rat and human pancreatic islets, increased insulin synthesis in purified rat islets, and caused a dose-dependent increase in plasma insulin levels in fasted rats, suggesting that VPAC2 receptor agonists may be a useful therapy for the treatment of type 2 diabetes [125].

There is persuasive evidence that VIP and PACAP play important roles in the control of immunity and inflammation (for review see [93]). PAC1 receptor mRNA is constitutively expressed in macrophages and monocytes. PACAP, acting through the PAC1 receptor appears to be protective against endotoxin-induced septic shock, acting at least in part by attenuating lipopolysaccharide-induced production of proinflammatory interleukin-6 [71]. VIP has potent effects in the immune system, influencing T cell differentiation and migration and modulating the production of cytokines by the two subsets of mouse helper T cells: T helper 1 (Th1) cells, which mediate classical delayed-type cellular immunity and T helper 2 (Th2) cells, which mediate hypersensitivity reactions, such as allergy. VPAC1 receptors are highly expressed constitutively on T cells, especially Th cells, whereas VPAC2 receptors are expressed marginally or not at all by unstimulated Th cells but are up-regulated to high levels by Th cell stimulation. Studies on VPAC2 receptor knockout mice [24,138] and on transgenic mice overexpressing the VPAC2 receptor in CD4 T cells [138-140] suggest that the receptor regulates the balance between Th1 and Th2 by stimulating production of more Th2-type cytokines, due to expansion of the Th2-type subset. PACAP knockout mice exhibited the predicted hyperinflammatory response in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, with an enhanced Th1/Th2 cytokine profile, but also with a reduced expansion of regulatory T cells [119]. VIP-deficient mice, on the other hand, exhibited a paradoxical resistance to EAE, with a failed entry of inflammatory cells into the CNS parenchyma [2], pointing to a critical role for VIP in T cell trafficking. VIP receptor agonists and antagonists may have therapeutic potential in the treatment of inflammatory and autoimmune diseases such as Crohn's disease [1], rheumatoid arthritis [54] and multiple sclerosis [26].

Studies on PAC1 receptor knockout mice point to a role for presynaptic PAC1-mediated signalling at the mossy fibre synapse in long-term potentiation (LTP) and hippocampus-dependent associative learning [72,86]. The PAC1 receptor is also expressed in brain areas which have been implicated in the emotional control of behaviour, such as the amygdala, bed nucleus of the stria terminalis (BNST), hypothalamus, locus coeruleus and periaqueductal grey. Consistent with this, PACAP and PAC1 receptors are upregulated in the BNST following chronic stress, and heightened BNST PACAP signalling produces anxiogenic behavioural responses [36]. PACAP and PAC1 receptor null mice demonstrate reduced anxiety behaviour and mice with a ubiquitous but not with a forebrain-specific deletion of the PAC1 receptor exhibited elevated locomotor activity with strongly reduced anxiety-like behaviour [72,86]. Furthermore, the glucocorticoid response in PACAP null animals is altered after emotional stressors [116,124]. PAC1 receptor signalling in the CNS also alters feeding behaviour [44,77]; PAC1 signalling decreases food intake and promotes anorexic-like responses, which may be related to enhanced anxiety. A polymorphism in the PAC1 receptor has been associated with post-traumatic stress disorder (PTSD) in the female population [99].

There is also clear evidence that PACAP exerts neurotrophic activities during development and may prevent brain damage provoked by various types of injury. PACAP and its receptors are expressed actively in the CNS during development [7-8,81]. In particular, high concentrations of PAC1 receptors are found in the external granule cell layer of the cerebellum during the two postnatal weeks [9,148], a period of intense multiplication and migration of granule cells. Treatment of cultured granule cells with PACAP enhances cell survival and stimulates neurite outgrowth [12,25,57]. The neurotrophic effect of PACAP is mediated through two distinct mechanisms, i.e. activation of the adenylyl cyclase and phospholipase C pathways leads to inhibition of caspase-3 activity and promotion of cell survival [132], whereas activation of the adenylyl cyclase and MAP-kinase pathways regulates gene expression and causes differentiation of granule neurons [130-131,137]. Injection for PACAP at the surface of the cerebellum of rat pups augments the number of migrating granule cells and increases the thickness of the internal granule cell layer [131], suggesting that PACAP is a potent inhibitor of apoptosis in the cerebellum during the development. Recent studies have shown that PACAP reduces the volume of infarct in a model of focal cerebral ischemia [97]. In vitro PACAP also exerts a neuroprotective effect on cerebellar neurons against apoptotic cell death induced by ethanol [135], ceramides [129] and oxidative stress [134].

The presence of PACAP in primary sensory neurones and the PAC1 receptor in the dorsal horn of the spinal cord [51] suggested a role for the PAC1 receptor in pain responses. PAC1 receptor knockout mice displayed impaired nociceptive responses to chemical, thermal and mechanical stimuli [52] and PACAP deficient mice also displayed abnormal pain responses [68].

Although most studies of PAC1 receptor knockout mice have found these animals to be superficially normal and viable, it has been reported that when crossed onto a C57BL/6 background, almost all PAC1 receptor knockout mice developed pulmonary hypertension and right heart failure after birth, suggesting an important role for PAC1-mediated signalling for the maintenance of normal pulmonary vascular tone during early postnatal life [85].

VIP is also thought to play a role in neurodevelopment and in neuroprotection following injury to the CNS. For example, VIP has been shown to be protective against excitotoxin-induced white matter lesions in neonatal mice [33-34,95], probably acting through VPAC2 receptors. VPAC2 receptors have also been implicated in the control of astrocyte proliferation [149]. NAP (davunetide), an active fragment of the VIP-inducted neuroprotective protein ADNP (activity-dependent neuroprotective protein) is in clinical development for the treatment of neurodegenerative disorders [30]. In studies of postnatal hippocampus in vitro, VPAC2 receptor activation was found to expand the pool of neural stem/progenitor cells by preventing either a neuronal or glial fate choice and by supporting their survival, whereas selective VPAC1 receptor activation promoted a neurogenic granule cell fate [147]. Two recent publications from independent groups have found associations between copy number variation in the gene encoding the VPAC2 receptor and susceptibility to schizophrenia [65,127]. These findings have generated some excitement in the field because they may imply that the VPAC2 receptor is a potential target for the development of new antipsychotic drugs [90].

Other receptors

There is some evidence for the presence of PHI-selective receptors in mammalian tissues [64,78] and the cloning of a PHI-selective receptor from the goldfish Carassius auratus has been reported [123]. Although there is no evidence at present for the existence of a separate gene encoding a PHI receptor in mammals [22], it remains possible that mammalian receptors with novel pharmacology, encoded by novel genes, resulting from alternative splicing of known genes or by interaction of known genes with accessory proteins, may be discovered in the future.

References

Show »

1. Abad C, Martinez C, Juarranz MG, Arranz A, Leceta J, Delgado M, Gomariz RP. (2003) Therapeutic effects of vasoactive intestinal peptide in the trinitrobenzene sulfonic acid mice model of Crohn's disease. Gastroenterology, 124: 961-971. [PMID:12671893]

2. Abad C, Tan YV, Lopez R, Nobuta H, Dong H, Phan P, Feng JM, Campagnoni AT, Waschek JA. (2010) Vasoactive intestinal peptide loss leads to impaired CNS parenchymal T-cell infiltration and resistance to experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA, 107 (45): 19555-60. [PMID:20978211]

3. Adamou JE, Aiyar N, Van Horn S, Elshourbagy NA. (1995) Cloning and functional characterization of the human vasoactive intestinal peptide (VIP)-2 receptor. Biochem Biophys Res Commun, 209 (2): 385-92. [PMID:7733904]

4. Arimura A, Shioda S. (1995) Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors: neuroendocrine and endocrine interaction. Front Neuroendocrinol, 16 (1): 53-88. [PMID:7768322]

5. Asnicar MA, Köster A, Heiman ML, Tinsley F, Smith DP, Galbreath E, Fox N, Ma YL, Blum WF, Hsiung HM. (2002) Vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating peptide receptor 2 deficiency in mice results in growth retardation and increased basal metabolic rate. Endocrinology, 143 (10): 3994-4006. [PMID:12239111]

6. Aton SJ, Colwell CS, Harmar AJ, Waschek J, Herzog ED. (2005) Vasoactive intestinal polypeptide mediates circadian rhythmicity and synchrony in mammalian clock neurons. Nat Neurosci, 8 (4): 476-83. [PMID:15750589]

7. Basille M, Gonzalez BJ, Fournier A, Vaudry H. (1994) Ontogeny of pituitary adenylate cyclase-activating polypeptide (PACAP) receptors in the rat cerebellum: a quantitative autoradiographic study. Brain Res Dev Brain Res, 82 (1-2): 81-9. [PMID:7842522]

8. Basille M, Gonzalez BJ, Leroux P, Jeandel L, Fournier A, Vaudry H. (1993) Localization and characterization of PACAP receptors in the rat cerebellum during development: evidence for a stimulatory effect of PACAP on immature cerebellar granule cells. Neuroscience, 57 (2): 329-38. [PMID:8115042]

9. Basille M, Vaudry D, Coulouarn Y, Jegou S, Lihrmann I, Fournier A, Vaudry H, Gonzalez B. (2000) Comparative distribution of pituitary adenylate cyclase-activating polypeptide (PACAP) binding sites and PACAP receptor mRNAs in the rat brain during development. J Comp Neurol, 425: 495-509. [PMID:10975876]

10. Besson J, Sarrieau A, Vial M, Marie JC, Rosselin G, Rostene W. (1986) Characterization and autoradiographic distribution of vasoactive intestinal peptide binding sites in the rat central nervous system. Brain Res, 398 (2): 329-36. [PMID:3801907]

11. Brenneman DE, Eiden LE. (1986) Vasoactive intestinal peptide and electrical activity influence neuronal survival. Proc Natl Acad Sci USA, 83 (4): 1159-62. [PMID:3456568]

12. Cavallaro S, Copani A, D'Agata V, Musco S, Petralia S, Ventra C, Stivala F, Travali S, Canonico PL. (1996) Pituitary adenylate cyclase activating polypeptide prevents apoptosis in cultured cerebellar granule neurons. Mol Pharmacol, 50 (1): 60-6. [PMID:8700120]

13. Colwell CS, Michel S, Itri J, Rodriguez W, Tam J, Lelievre V, Hu Z, Liu X, Waschek JA. (2003) Disrupted circadian rhythms in VIP- and PHI-deficient mice. Am J Physiol Regul Integr Comp Physiol, 285 (5): R939-49. [PMID:12855416]

14. Colwell CS, Michel S, Itri J, Rodriguez W, Tam J, Lelièvre V, Hu Z, Waschek JA. (2004) Selective deficits in the circadian light response in mice lacking PACAP. Am J Physiol Regul Integr Comp Physiol, 287 (5): R1194-201. [PMID:15217792]

15. Couvineau A, Laburthe M. (2012) The family B1 GPCR: structural aspects and interaction with accessory proteins. Curr Drug Targets, 13 (1): 103-15. [PMID:21777182]

16. Couvineau A, Rouyer-Fessard C, Maoret JJ, Gaudin P, Nicole P, Laburthe M. (1996) Vasoactive intestinal peptide (VIP)1 receptor. Three nonadjacent amino acids are responsible for species selectivity with respect to recognition of peptide histidine isoleucineamide. J Biol Chem, 271 (22): 12795-800. [PMID:8662697]

17. Cutler DJ, Haraura M, Reed HE, Shen S, Sheward WJ, Morrison CF, Marston HM, Harmar AJ, Piggins HD. (2003) The mouse VPAC2 receptor confers suprachiasmatic nuclei cellular rhythmicity and responsiveness to vasoactive intestinal polypeptide in vitro. Eur J Neurosci, 17 (2): 197-204. [PMID:12542655]

18. Daniel PB, Kieffer TJ, Leech CA, Habener JF. (2001) Novel alternatively spliced exon in the extracellular ligand-binding domain of the pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1R) selectively increases ligand affinity and alters signal transduction coupling during spermatogenesis. J Biol Chem, 276 (16): 12938-44. [PMID:11278585]

19. Dautzenberg FM, Mevenkamp G, Wille S, Hauger RL. (1999) N-terminal splice variants of the type I PACAP receptor: isolation, characterization and ligand binding/selectivity determinants. J Neuroendocrinol, 11 (12): 941-9. [PMID:10583729]

20. Delgado M, Martinez C, Johnson MC, Gomariz RP, Ganea D. (1996) Differential expression of vasoactive intestinal peptide receptors 1 and 2 (VIP-R1 and VIP-R2) mRNA in murine lymphocytes. J Neuroimmunol, 68 (1-2): 27-38. [PMID:8784257]

21. Fabricius D, Karacay B, Shutt D, Leverich W, Schafer B, Takle E, Thedens D, Khanna G, Raikwar S, Yang B et al.. (2011) Characterization of intestinal and pancreatic dysfunction in VPAC1-null mutant mouse. Pancreas, 40 (6): 861-71. [PMID:21697765]

22. Foord SM, Bonner TI, Neubig RR, Rosser EM, Pin JP, Davenport AP, Spedding M, Harmar AJ. (2005) International Union of Pharmacology. XLVI. G protein-coupled receptor list. Pharmacol Rev, 57 (2): 279-88. [PMID:15914470]

23. Ghatei MA, Takahashi K, Suzuki Y, Gardiner J, Jones PM, Bloom SR. (1993) Distribution, molecular characterization of pituitary adenylate cyclase-activating polypeptide and its precursor encoding messenger RNA in human and rat tissues. J Endocrinol, 136 (1): 159-66. [PMID:8094091]

24. Goetzl EJ, Voice JK, Shen S, Dorsam G, Kong Y, West KM, Morrison CF, Harmar AJ. (2001) Enhanced delayed-type hypersensitivity and diminished immediate-type hypersensitivity in mice lacking the inducible VPAC(2) receptor for vasoactive intestinal peptide. Proc Natl Acad Sci USA, 98 (24): 13854-9. [PMID:11698667]

25. Gonzalez BJ, Basille M, Vaudry D, Fournier A, Vaudry H. (1997) Pituitary adenylate cyclase-activating polypeptide promotes cell survival and neurite outgrowth in rat cerebellar neuroblasts. Neuroscience, 78 (2): 419-30. [PMID:9145799]

26. Gonzalez-Rey E, Fernandez-Martin A, Chorny A, Martin J, Pozo D, Ganea D, Delgado M. (2006) Therapeutic effect of vasoactive intestinal peptide on experimental autoimmune encephalomyelitis: down-regulation of inflammatory and autoimmune responses. Am J Pathol, 168 (4): 1179-88. [PMID:16565493]

27. Gourlet P, De Neef P, Cnudde J, Waelbroeck M, Robberecht P. (1997) In vitro properties of a high affinity selective antagonist of the VIP1 receptor. Peptides, 18 (10): 1555-60. [PMID:9437716]

28. Gourlet P, Vandermeers A, Vertongen P, Rathe J, De Neef P, Cnudde J, Waelbroeck M, Robberecht P. (1997) Development of high affinity selective VIP1 receptor agonists. Peptides, 18 (10): 1539-45. [PMID:9437714]

29. Gourlet P, Vertongen P, Vandermeers A, Vandermeers-Piret MC, Rathe J, De Neef P, Waelbroeck M, Robberecht P. (1997) The long-acting vasoactive intestinal polypeptide agonist RO 25-1553 is highly selective of the VIP2 receptor subclass. Peptides, 18 (3): 403-8. [PMID:9145428]

30. Gozes I. (2011) NAP (davunetide) provides functional and structural neuroprotection. Curr Pharm Des, 17 (10): 1040-4. [PMID:21524250]

31. Gozes I, Brenneman DE. (1989) VIP: molecular biology and neurobiological function. Mol Neurobiol, 3 (4): 201-36. [PMID:2698176]

32. Grace CR, Perrin MH, DiGruccio MR, Miller CL, Rivier JE, Vale WW, Riek R. (2004) NMR structure and peptide hormone binding site of the first extracellular domain of a type B1 G protein-coupled receptor. Proc Natl Acad Sci USA, 101 (35): 12836-41. [PMID:15326300]

33. Gressens P, Besse L, Robberecht P, Gozes I, Fridkin M, Evrard P. (1999) Neuroprotection of the developing brain by systemic administration of vasoactive intestinal peptide derivatives. J Pharmacol Exp Ther, 288 (3): 1207-13. [PMID:10027860]

34. Gressens P, Marret S, Hill JM, Brenneman DE, Gozes I, Fridkin M, Evrard P. (1997) Vasoactive intestinal peptide prevents excitotoxic cell death in the murine developing brain. J Clin Invest, 100 (2): 390-7. [PMID:9218516]

35. Hamelink C, Tjurmina O, Damadzic R, Young WS, Weihe E, Lee HW, Eiden LE. (2002) Pituitary adenylate cyclase-activating polypeptide is a sympathoadrenal neurotransmitter involved in catecholamine regulation and glucohomeostasis. Proc Natl Acad Sci USA, 99 (1): 461-6. [PMID:11756684]

36. Hammack SE, Roman CW, Lezak KR, Kocho-Shellenberg M, Grimmig B, Falls WA, Braas K, May V. (2010) Roles for pituitary adenylate cyclase-activating peptide (PACAP) expression and signaling in the bed nucleus of the stria terminalis (BNST) in mediating the behavioral consequences of chronic stress. J Mol Neurosci, 42 (3): 327-40. [PMID:20405238]

37. Hannibal J, Ding JM, Chen D, Fahrenkrug J, Larsen PJ, Gillette MU, Mikkelsen JD. (1997) Pituitary adenylate cyclase-activating peptide (PACAP) in the retinohypothalamic tract: a potential daytime regulator of the biological clock. J Neurosci, 17 (7): 2637-44. [PMID:9065523]

38. Hannibal J, Jamen F, Nielsen HS, Journot L, Brabet P, Fahrenkrug J. (2001) Dissociation between light-induced phase shift of the circadian rhythm and clock gene expression in mice lacking the pituitary adenylate cyclase activating polypeptide type 1 receptor. J Neurosci, 21 (13): 4883-90. [PMID:11425915]

39. Harmar AJ. (2001) Family-B G-protein-coupled receptors. Genome Biol, 2 (12): REVIEWS3013. [PMID:11790261]

40. Harmar AJ, Arimura A, Gozes I, Journot L, Laburthe M, Pisegna JR, Rawlings SR, Robberecht P, Said SI, Sreedharan SP et al.. (1998) International Union of Pharmacology. XVIII. Nomenclature of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Pharmacol Rev, 50 (2): 265-70. [PMID:9647867]

41. Harmar AJ, Marston HM, Shen S, Spratt C, West KM, Sheward WJ, Morrison CF, Dorin JR, Piggins HD, Reubi JC et al.. (2002) The VPAC(2) receptor is essential for circadian function in the mouse suprachiasmatic nuclei. Cell, 109 (4): 497-508. [PMID:12086606]

42. Harmar AJ, Sheward WJ, Morrison CF, Waser B, Gugger M, Reubi JC. (2004) Distribution of the VPAC2 receptor in peripheral tissues of the mouse. Endocrinology, 145 (3): 1203-10. [PMID:14617572]

43. Hashimoto H, Nogi H, Mori K, Ohishi H, Shigemoto R, Yamamoto K, Matsuda T, Mizuno N, Nagata S, Baba A. (1996) Distribution of the mRNA for a pituitary adenylate cyclase-activating polypeptide receptor in the rat brain: an in situ hybridization study. J Comp Neurol, 371 (4): 567-77. [PMID:8841910]

44. Hawke Z, Ivanov TR, Bechtold DA, Dhillon H, Lowell BB, Luckman SM. (2009) PACAP neurons in the hypothalamic ventromedial nucleus are targets of central leptin signaling. J Neurosci, 29 (47): 14828-35. [PMID:19940178]

45. Hughes AT, Fahey B, Cutler DJ, Coogan AN, Piggins HD. (2004) Aberrant gating of photic input to the suprachiasmatic circadian pacemaker of mice lacking the VPAC2 receptor. J Neurosci, 24 (14): 3522-6. [PMID:15071099]

46. Ichikawa S, Sreedharan SP, Owen RL, Goetzl EJ. (1995) Immunochemical localization of type I VIP receptor and NK-1-type substance P receptor in rat lung. Am J Physiol, 268 (4 Pt 1): L584-8. [PMID:7537460]

47. Inagaki N, Yoshida H, Mizuta M, Mizuno N, Fujii Y, Gonoi T, Miyazaki J, Seino S. (1994) Cloning and functional characterization of a third pituitary adenylate cyclase-activating polypeptide receptor subtype expressed in insulin-secreting cells. Proc Natl Acad Sci USA, 91 (7): 2679-83. [PMID:8146174]

48. Ishihara T, Shigemoto R, Mori K, Takahashi K, Nagata S. (1992) Functional expression and tissue distribution of a novel receptor for vasoactive intestinal polypeptide. Neuron, 8: 811-819. [PMID:1314625]

49. Itoh N, Obata K, Yanaihara N, Okamoto H. (1983) Human preprovasoactive intestinal polypeptide contains a novel PHI-27-like peptide, PHM-27. Nature, 304 (5926): 547-9. [PMID:6571696]

50. Jamen F, Persson K, Bertrand G, Rodriguez-Henche N, Puech R, Bockaert J, Ahrén B, Brabet P. (2000) PAC1 receptor-deficient mice display impaired insulinotropic response to glucose and reduced glucose tolerance. J Clin Invest, 105 (9): 1307-15. [PMID:10792006]

51. Jongsma H, Danielsen N, Sundler F, Kanje M. (2000) Alteration of PACAP distribution and PACAP receptor binding in the rat sensory nervous system following sciatic nerve transection. Brain Res, 853: 186-196. [PMID:10640616]

52. Jongsma H, Pettersson LM, Zhang Yz, Reimer MK, Kanje M, Waldenström A, Sundler F, Danielsen N. (2001) Markedly reduced chronic nociceptive response in mice lacking the PAC1 receptor. Neuroreport, 12 (10): 2215-9. [PMID:11447337]

53. Journot L, Waeber C, Pantaloni C, Holsboer F, Seeburg PH, Bockaert J, Spengler D. (1995) Differential signal transduction by six splice variants of the pituitary adenylate cyclase-activating peptide (PACAP) receptor. Biochem Soc Trans, 23: 133-137. [PMID:7758697]

54. Juarranz MG, Santiago B, Torroba M, Gutierrez-Cañas I, Palao G, Galindo M, Abad C, Martinez C, Leceta J, Pablos JL et al.. (2004) Vasoactive intestinal peptide modulates proinflammatory mediator synthesis in osteoarthritic and rheumatoid synovial cells. Rheumatology (Oxford), 43 (4): 416-22. [PMID:14657510]

55. Kaltreider HB, Ichikawa S, Byrd PK, Ingram DA, Kishiyama JL, Sreedharan SP, Warnock ML, Beck JM, Goetzl EJ. (1997) Upregulation of neuropeptides and neuropeptide receptors in a murine model of immune inflammation in lung parenchyma. Am J Respir Cell Mol Biol, 16 (2): 133-44. [PMID:9032120]

56. Kawaguchi C, Tanaka K, Isojima Y, Shintani N, Hashimoto H, Baba A, Nagai K. (2003) Changes in light-induced phase shift of circadian rhythm in mice lacking PACAP. Biochem Biophys Res Commun, 310 (1): 169-75. [PMID:14511666]

57. Kienlen Campard P, Crochemore C, René F, Monnier D, Koch B, Loeffler JP. (1997) PACAP type I receptor activation promotes cerebellar neuron survival through the cAMP/PKA signaling pathway. DNA Cell Biol, 16 (3): 323-33. [PMID:9115641]

58. Krempels K, Usdin TB, Harta G, Mezey E. (1995) PACAP acts through VIP type 2 receptors in the rat testis. Neuropeptides, 29 (6): 315-20. [PMID:8837957]

59. Kumar S, Pioszak A, Zhang C, Swaminathan K, Xu HE. (2011) Crystal Structure of the PAC1R Extracellular Domain Unifies a Consensus Fold for Hormone Recognition by Class B G-Protein Coupled Receptors. PLoS ONE, 6 (5): e19682. [PMID:21625560]

60. Laburthe M, Amiranoff B, Boige N, Rouyer-Fessard C, Tatemoto K, Moroder L. (1983) Interaction of GRF with VIP receptors and stimulation of adenylate cyclase in rat and human intestinal epithelial membranes. Comparison with PHI and secretin. FEBS Lett, 159 (1-2): 89-92. [PMID:6307754]

61. Laburthe M, Couvineau A. (2002) Molecular pharmacology and structure of VPAC Receptors for VIP and PACAP. Regul Pept, 108 (2-3): 165-73. [PMID:12220741]

62. Laburthe M, Couvineau A, Marie JC. (2002) VPAC receptors for VIP and PACAP. Recept Channels, 8 (3-4): 137-53. [PMID:12529932]

63. Laburthe M, Couvineau A, Tan V. (2007) Class II G protein-coupled receptors for VIP and PACAP: structure, models of activation and pharmacology. Peptides, 28 (9): 1631-9. [PMID:17574305]

64. Lelièvre V, Pineau N, Du J, Wen CH, Nguyen T, Janet T, Muller JM, Waschek JA. (1998) Differential effects of peptide histidine isoleucine (PHI) and related peptides on stimulation and suppression of neuroblastoma cell proliferation. A novel VIP-independent action of PHI via MAP kinase. J Biol Chem, 273 (31): 19685-90. [PMID:9677397]

65. Levinson DF, Duan J, Oh S, Wang K, Sanders AR, Shi J, Zhang N, Mowry BJ, Olincy A, Amin F et al.. (2011) Copy number variants in schizophrenia: confirmation of five previous findings and new evidence for 3q29 microdeletions and VIPR2 duplications. Am J Psychiatry, 168 (3): 302-16. [PMID:21285140]

66. Luis J, Said SI. (1990) Characterization of VIP- and helodermin-preferring receptors on human small cell lung carcinoma cell lines. Peptides, 11 (6): 1239-44. [PMID:1965034]

67. Lutz EM, Sheward WJ, West KM, Morrow JA, Fink G, Harmar AJ. (1993) The VIP2 receptor: molecular characterisation of a cDNA encoding a novel receptor for vasoactive intestinal peptide. FEBS Lett, 334 (1): 3-8. [PMID:8224221]

68. Mabuchi T, Shintani N, Matsumura S, Okuda-Ashitaka E, Hashimoto H, Muratani T, Minami T, Baba A, Ito S. (2004) Pituitary adenylate cyclase-activating polypeptide is required for the development of spinal sensitization and induction of neuropathic pain. J Neurosci, 24 (33): 7283-91. [PMID:15317855]

69. Malhotra RK, Wakade TD, Wakade AR. (1988) Vasoactive intestinal polypeptide and muscarine mobilize intracellular Ca2+ through breakdown of phosphoinositides to induce catecholamine secretion. Role of IP3 in exocytosis. J Biol Chem, 263 (5): 2123-6. [PMID:3123488]

70. Martin JL, Dietl MM, Hof PR, Palacios JM, Magistretti PJ. (1987) Autoradiographic mapping of [mono[125I]iodo-Tyr10, MetO17]vasoactive intestinal peptide binding sites in the rat brain. Neuroscience, 23 (2): 539-65. [PMID:3437978]

71. Martinez C, Abad C, Delgado M, Arranz A, Juarranz MG, Rodriguez-Henche N, Brabet P, Leceta J, Gomariz RP. (2002) Anti-inflammatory role in septic shock of pituitary adenylate cyclase-activating polypeptide receptor. Proc Natl Acad Sci USA, 99 (2): 1053-8. [PMID:11792830]

72. Matsuyama S, Matsumoto A, Hashimoto H, Shintani N, Baba A. (2003) Impaired long-term potentiation in vivo in the dentate gyrus of pituitary adenylate cyclase-activating polypeptide (PACAP) or PACAP type 1 receptor-mutant mice. Neuroreport, 14 (16): 2095-8. [PMID:14600504]

73. Miyata A, Arimura A, Dahl RR, Minamino N, Uehara A, Jiang L, Culler MD, Coy DH. (1989) Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun, 164 (1): 567-74. [PMID:2803320]

74. Miyata A, Jiang L, Dahl RD, Kitada C, Kubo K, Fujino M, Minamino N, Arimura A. (1990) Isolation of a neuropeptide corresponding to the N-terminal 27 residues of the pituitary adenylate cyclase activating polypeptide with 38 residues (PACAP38). Biochem Biophys Res Commun, 170 (2): 643-8. [PMID:2383262]

75. Moller K, Sundler F. (1996) Expression of pituitary adenylate cyclase activating peptide (PACAP) and PACAP type I receptors in the rat adrenal medulla. Regul Pept, 63 (2-3): 129-39. [PMID:8837221]

76. Moreno D, Gourlet P, De Neef P, Cnudde J, Waelbroeck M, Robberecht P. (2000) Development of selective agonists and antagonists for the human vasoactive intestinal polypeptide VPAC(2) receptor. Peptides, 21 (10): 1543-9. [PMID:11068102]

77. Mounien L, Do Rego JC, Bizet P, Boutelet I, Gourcerol G, Fournier A, Brabet P, Costentin J, Vaudry H, Jégou S. (2009) Pituitary adenylate cyclase-activating polypeptide inhibits food intake in mice through activation of the hypothalamic melanocortin system. Neuropsychopharmacology, 34 (2): 424-35. [PMID:18536705]

78. Murthy KS, Zhang KM, Jin JG, Grider JR, Makhlouf GM. (1993) VIP-mediated G protein-coupled Ca2+ influx activates a constitutive NOS in dispersed gastric muscle cells. Am J Physiol, 265 (4 Pt 1): G660-71. [PMID:7694477]

79. Neumann JM, Couvineau A, Murail S, Lacapère JJ, Jamin N, Laburthe M. (2008) Class-B GPCR activation: is ligand helix-capping the key?. Trends Biochem Sci, 33 (7): 314-9. [PMID:18555686]

80. Nicole P, Lins L, Rouyer-Fessard C, Drouot C, Fulcrand P, Thomas A, Couvineau A, Martinez J, Brasseur R, Laburthe M. (2000) Identification of key residues for interaction of vasoactive intestinal peptide with human VPAC1 and VPAC2 receptors and development of a highly selective VPAC1 receptor agonist. Alanine scanning and molecular modeling of the peptide. J Biol Chem, 275 (31): 24003-12. [PMID:10801840]

81. Nielsen HS, Hannibal J, Fahrenkrug J. (1998) Expression of pituitary adenylate cyclase activating polypeptide (PACAP) in the postnatal and adult rat cerebellar cortex. Neuroreport, 9 (11): 2639-42. [PMID:9721947]

82. O'Donnell M, Garippa RJ, Rinaldi N, Selig WM, Simko B, Renzetti L, Tannu SA, Wasserman MA, Welton A, Bolin DR. (1994) Ro 25-1553: a novel, long-acting vasoactive intestinal peptide agonist. Part I: In vitro and in vivo bronchodilator studies. J Pharmacol Exp Ther, 270 (3): 1282-8. [PMID:7932180]

83. O'Donnell M, Garippa RJ, Rinaldi N, Selig WM, Tocker JE, Tannu SA, Wasserman MA, Welton A, Bolin DR. (1994) Ro 25-1553: a novel, long-acting vasoactive intestinal peptide agonist. Part II: Effect on in vitro and in vivo models of pulmonary anaphylaxis. J Pharmacol Exp Ther, 270 (3): 1289-94. [PMID:7932181]

84. Ottaway CA. (1987) Selective effects of vasoactive intestinal peptide on the mitogenic response of murine T cells. Immunology, 62 (2): 291-7. [PMID:2960611]

85. Otto C, Hein L, Brede M, Jahns R, Engelhardt S, Gröne HJ, Schütz G. (2004) Pulmonary hypertension and right heart failure in pituitary adenylate cyclase-activating polypeptide type I receptor-deficient mice. Circulation, 110 (20): 3245-51. [PMID:15520307]

86. Otto C, Kovalchuk Y, Wolfer DP, Gass P, Martin M, Zuschratter W, Gröne HJ, Kellendonk C, Tronche F, Maldonado R et al.. (2001) Impairment of mossy fiber long-term potentiation and associative learning in pituitary adenylate cyclase activating polypeptide type I receptor-deficient mice. J Neurosci, 21 (15): 5520-7. [PMID:11466423]

87. Otto C, Martin M, Wolfer DP, Lipp HP, Maldonado R, Schütz G. (2001) Altered emotional behavior in PACAP-type-I-receptor-deficient mice. Brain Res Mol Brain Res, 92 (1-2): 78-84. [PMID:11483244]

88. Pantaloni C, Brabet P, Bilanges B, Dumuis A, Houssami S, Spengler D, Bockaert J, Journot L. (1996) Alternative splicing in the N-terminal extracellular domain of the pituitary adenylate cyclase-activating polypeptide (PACAP) receptor modulates receptor selectivity and relative potencies of PACAP-27 and PACAP-38 in phospholipase C activation. J Biol Chem, 271 (36): 22146-51. [PMID:8703026]

89. Persson K, Ahrén B. (2002) The neuropeptide PACAP contributes to the glucagon response to insulin-induced hypoglycaemia in mice. Acta Physiol Scand, 175 (1): 25-8. [PMID:11982501]

90. Piggins HD. (2011) Schizophrenia: Zooming in on a gene. Nature, 471 (7339): 455-6. [PMID:21430769]

91. Pisegna JR, Wank SA. (1993) Molecular cloning and functional expression of the pituitary adenylate cyclase-activating polypeptide type I receptor. Proc Natl Acad Sci USA, 90 (13): 6345-9. [PMID:8392197]

92. Pisegna JR, Wank SA. (1996) Cloning and characterization of the signal transduction of four splice variants of the human pituitary adenylate cyclase activating polypeptide receptor. Evidence for dual coupling to adenylate cyclase and phospholipase C. J Biol Chem, 271 (29): 17267-74. [PMID:8663363]

93. Pozo D. (2003) VIP- and PACAP-mediated immunomodulation as prospective therapeutic tools. Trends Mol Med, 9 (5): 211-7. [PMID:12763526]

94. Przywara DA, Guo X, Angelilli ML, Wakade TD, Wakade AR. (1996) A non-cholinergic transmitter, pituitary adenylate cyclase-activating polypeptide, utilizes a novel mechanism to evoke catecholamine secretion in rat adrenal chromaffin cells. J Biol Chem, 271 (18): 10545-50. [PMID:8631854]

95. Rangon CM, Goursaud S, Medja F, Lelièvre V, Mounien L, Husson I, Brabet P, Jégou S, Janet T, Gressens P. (2005) VPAC2 receptors mediate vasoactive intestinal peptide-induced neuroprotection against neonatal excitotoxic brain lesions in mice. J Pharmacol Exp Ther, 314 (2): 745-52. [PMID:15872042]

96. Rawlings SR, Hezareh M. (1996) Pituitary adenylate cyclase-activating polypeptide (PACAP) and PACAP/vasoactive intestinal polypeptide receptors: actions on the anterior pituitary gland. Endocr Rev, 17 (1): 4-29. [PMID:8641222]

97. Reglodi D, Somogyvari-Vigh A, Vigh S, Kozicz T, Arimura A. (2000) Delayed systemic administration of PACAP38 is neuroprotective in transient middle cerebral artery occlusion in the rat. Stroke, 31 (6): 1411-7. [PMID:10835464]

98. Reichlin S. (1988) Neuroendocrine significance of vasoactive intestinal polypeptide. Ann N Y Acad Sci, 527: 431-49. [PMID:2898911]

99. Ressler KJ, Mercer KB, Bradley B, Jovanovic T, Mahan A, Kerley K, Norrholm SD, Kilaru V, Smith AK, Myers AJ et al.. (2011) Post-traumatic stress disorder is associated with PACAP and the PAC1 receptor. Nature, 470 (7335): 492-7. [PMID:21350482]

100. Reubi JC. (2000) In vitro evaluation of VIP/PACAP receptors in healthy and diseased human tissues. Clinical implications. Ann N Y Acad Sci, 921: 1-25. [PMID:11193811]

101. Reubi JC, Läderach U, Waser B, Gebbers JO, Robberecht P, Laissue JA. (2000) Vasoactive intestinal peptide/pituitary adenylate cyclase-activating peptide receptor subtypes in human tumors and their tissues of origin. Cancer Res, 60 (11): 3105-12. [PMID:10850463]

102. Robberecht P, Waelbroeck M, De Neef P, Tastenoy M, Gourlet P, Cogniaux J, Christophe J. (1988) A new type of functional VIP receptor has an affinity for helodermin in human SUP-T1 lymphoblasts. FEBS Lett, 228 (2): 351-5. [PMID:2830146]

103. Robberecht P, Woussen-Colle MC, De Neef P, Gourlet P, Buscail L, Vandermeers A, Vandermeers-Piret MC, Christophe J. (1991) The two forms of the pituitary adenylate cyclase activating polypeptide (PACAP (1-27) and PACAP (1-38)) interact with distinct receptors on rat pancreatic AR 4-2J cell membranes. FEBS Lett, 286 (1-2): 133-6. [PMID:1650711]

104. Said SI. (1991) Vasoactive intestinal polypeptide: biologic role in health and disease. Trends Endocrinol Metab, 2 (3): 107-12. [PMID:18411175]

105. Said SI. (1996) Vasoactive intestinal peptide and nitric oxide: divergent roles in relation to tissue injury. Ann N Y Acad Sci, 805: 379-87; discussion 387-8. [PMID:8993418]

106. Said SI, Mutt V. (1970) Polypeptide with broad biological activity: isolation from small intestine. Science, 169 (3951): 1217-8. [PMID:5450698]

107. Said SI, Mutt V. (1972) Isolation from porcine-intestinal wall of a vasoactive octacosapeptide related to secretin and to glucagon. Eur J Biochem, 28 (2): 199-204. [PMID:5069712]

108. Said SI, Rattan S. (2004) The multiple mediators of neurogenic smooth muscle relaxation. Trends Endocrinol Metab, 15 (5): 189-91. [PMID:15223046]

109. Sheward WJ, Lutz EM, Copp AJ, Harmar AJ. (1998) Expression of PACAP, and PACAP type 1 (PAC1) receptor mRNA during development of the mouse embryo. Brain Res Dev Brain Res, 109 (2): 245-53. [PMID:9729410]

110. Sheward WJ, Lutz EM, Harmar AJ. (1995) The distribution of vasoactive intestinal peptide2 receptor messenger RNA in the rat brain and pituitary gland as assessed by in situ hybridization. Neuroscience, 67 (2): 409-18. [PMID:7675176]

111. Shioda S, Shuto Y, Somogyvari-Vigh A, Legradi G, Onda H, Coy DH, Nakajo S, Arimura A. (1997) Localization and gene expression of the receptor for pituitary adenylate cyclase-activating polypeptide in the rat brain. Neurosci Res, 28 (4): 345-54. [PMID:9274830]

112. Shivers BD, Görcs TJ, Gottschall PE, Arimura A. (1991) Two high affinity binding sites for pituitary adenylate cyclase-activating polypeptide have different tissue distributions. Endocrinology, 128 (6): 3055-65. [PMID:2036976]

113. Sorg O, Magistretti PJ. (1992) Vasoactive intestinal peptide and noradrenaline exert long-term control on glycogen levels in astrocytes: blockade by protein synthesis inhibition. J Neurosci, 12 (12): 4923-31. [PMID:1334506]

114. Spengler D, Waeber C, Pantaloni C, Holsboer F, Bockaert J, Seeburg PH, Journot L. (1993) Differential signal transduction by five splice variants of the PACAP receptor. Nature, 365 (6442): 170-5. [PMID:8396727]

115. Sreedharan SP, Huang JX, Cheung MC, Goetzl EJ. (1995) Structure, expression, and chromosomal localization of the type I human vasoactive intestinal peptide receptor gene. Proc Natl Acad Sci USA, 92 (7): 2939-43. [PMID:7708752]

116. Stroth N, Eiden LE. (2010) Stress hormone synthesis in mouse hypothalamus and adrenal gland triggered by restraint is dependent on pituitary adenylate cyclase-activating polypeptide signaling. Neuroscience, 165 (4): 1025-30. [PMID:19931358]

117. Sun C, Song D, Davis-Taber RA, Barrett LW, Scott VE, Richardson PL, Pereda-Lopez A, Uchic ME, Solomon LR, Lake MR et al.. (2007) Solution structure and mutational analysis of pituitary adenylate cyclase-activating polypeptide binding to the extracellular domain of PAC1-RS. Proc Natl Acad Sci USA, 104 (19): 7875-80. [PMID:17470806]

118. Sundler F. (1999) Preliminary observations of phenotypic alterations in the PAC1 receptor knockout mouse. Soc Neurosci Abstracts, 25: 441-441.

119. Tan YV, Abad C, Lopez R, Dong H, Liu S, Lee A, Gomariz RP, Leceta J, Waschek JA. (2009) Pituitary adenylyl cyclase-activating polypeptide is an intrinsic regulator of Treg abundance and protects against experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA, 106 (6): 2012-7. [PMID:19190179]

120. Tan YV, Couvineau A, Murail S, Ceraudo E, Neumann JM, Lacapère JJ, Laburthe M. (2006) Peptide agonist docking in the N-terminal ectodomain of a class II G protein-coupled receptor, the VPAC1 receptor. Photoaffinity, NMR, and molecular modeling. J Biol Chem, 281 (18): 12792-8. [PMID:16520374]

121. Tatemoto K, Mutt V. (1981) Isolation and characterization of the intestinal peptide porcine PHI (PHI-27), a new member of the glucagon--secretin family. Proc Natl Acad Sci USA, 78 (11): 6603-7. [PMID:6947244]

122. Tomimoto S, Hashimoto H, Shintani N, Yamamoto K, Kawabata Y, Hamagami K, Yamagata K, Miyagawa J, Baba A. (2004) Overexpression of pituitary adenylate cyclase-activating polypeptide in islets inhibits hyperinsulinemia and islet hyperplasia in agouti yellow mice. J Pharmacol Exp Ther, 309 (2): 796-803. [PMID:14742740]

123. Tse DL, Pang RT, Wong AO, Chan SM, Vaudry H, Chow BK. (2002) Identification of a potential receptor for both peptide histidine isoleucine and peptide histidine valine. Endocrinology, 143 (4): 1327-36. [PMID:11897689]

124. Tsukiyama N, Saida Y, Kakuda M, Shintani N, Hayata A, Morita Y, Tanida M, Tajiri M, Hazama K, Ogata K et al.. (2011) PACAP centrally mediates emotional stress-induced corticosterone responses in mice. Stress, 14 (4): 368-75. [PMID:21438773]

125. Tsutsumi M, Claus TH, Liang Y, Li Y, Yang L, Zhu J, Dela Cruz F, Peng X, Chen H, Yung SL et al.. (2002) A potent and highly selective VPAC2 agonist enhances glucose-induced insulin release and glucose disposal: a potential therapy for type 2 diabetes. Diabetes, 51 (5): 1453-60. [PMID:11978642]

126. Usdin TB, Bonner TI, Mezey E. (1994) Two receptors for vasoactive intestinal polypeptide with similar specificity and complementary distributions. Endocrinology, 135 (6): 2662-80. [PMID:7988457]

127. Vacic V, McCarthy S, Malhotra D, Murray F, Chou HH, Peoples A, Makarov V, Yoon S, Bhandari A, Corominas R et al.. (2011) Duplications of the neuropeptide receptor gene VIPR2 confer significant risk for schizophrenia. Nature, 471 (7339): 499-503. [PMID:21346763]

128. Van Rampelbergh J, Gourlet P, De Neef P, Robberecht P, Waelbroeck M. (1996) Properties of the pituitary adenylate cyclase-activating polypeptide I and II receptors, vasoactive intestinal peptide1, and chimeric amino-terminal pituitary adenylate cyclase-activating polypeptide/vasoactive intestinal peptide1 receptors: evidence for multiple receptor states. Mol Pharmacol, 50 (6): 1596-604. [PMID:8967982]

129. Vaudry D, Falluel-Morel A, Basille M, Pamantung TF, Fontaine M, Fournier A, Vaudry H, Gonzalez BJ. (2003) Pituitary adenylate cyclase-activating polypeptide prevents C2-ceramide-induced apoptosis of cerebellar granule cells. J Neurosci Res, 72 (3): 303-16. [PMID:12692897]

130. Vaudry D, Gonzalez BJ, Basille M, Anouar Y, Fournier A, Vaudry H. (1998) Pituitary adenylate cyclase-activating polypeptide stimulates both c-fos gene expression and cell survival in rat cerebellar granule neurons through activation of the protein kinase A pathway. Neuroscience, 84 (3): 801-12. [PMID:9579785]

131. Vaudry D, Gonzalez BJ, Basille M, Fournier A, Vaudry H. (1999) Neurotrophic activity of pituitary adenylate cyclase-activating polypeptide on rat cerebellar cortex during development. Proc Natl Acad Sci USA, 96 (16): 9415-20. [PMID:10430957]

132. Vaudry D, Gonzalez BJ, Basille M, Pamantung TF, Fontaine M, Fournier A, Vaudry H. (2000) The neuroprotective effect of pituitary adenylate cyclase-activating polypeptide on cerebellar granule cells is mediated through inhibition of the CED3-related cysteine protease caspase-3/CPP32. Proc Natl Acad Sci USA, 97 (24): 13390-5. [PMID:11087878]

133. Vaudry D, Gonzalez BJ, Basille M, Yon L, Fournier A, Vaudry H. (2000) Pituitary adenylate cyclase-activating polypeptide and its receptors: from structure to functions. Pharmacol Rev, 52: 269-324. [PMID:10835102]

134. Vaudry D, Pamantung TF, Basille M, Rousselle C, Fournier A, Vaudry H, Beauvillain JC, Gonzalez BJ. (2002) PACAP protects cerebellar granule neurons against oxidative stress-induced apoptosis. Eur J Neurosci, 15 (9): 1451-60. [PMID:12028355]

135. Vaudry D, Rousselle C, Basille M, Falluel-Morel A, Pamantung TF, Fontaine M, Fournier A, Vaudry H, Gonzalez BJ. (2002) Pituitary adenylate cyclase-activating polypeptide protects rat cerebellar granule neurons against ethanol-induced apoptotic cell death. Proc Natl Acad Sci USA, 99 (9): 6398-403. [PMID:11972030]

136. Vertongen P, Schiffmann SN, Gourlet P, Robberecht P. (1997) Autoradiographic visualization of the receptor subclasses for vasoactive intestinal polypeptide (VIP) in rat brain. Peptides, 18: 1547-1554. [PMID:9437715]

137. Villalba M, Bockaert J, Journot L. (1997) Pituitary adenylate cyclase-activating polypeptide (PACAP-38) protects cerebellar granule neurons from apoptosis by activating the mitogen-activated protein kinase (MAP kinase) pathway. J Neurosci, 17 (1): 83-90. [PMID:8987738]

138. Voice JK, Dorsam G, Chan RC, Grinninger C, Kong Y, Goetzl EJ. (2002) Immunoeffector and immunoregulatory activities of vasoactive intestinal peptide. Regul Pept, 109 (1-3): 199-208. [PMID:12409234]

139. Voice JK, Dorsam G, Lee H, Kong Y, Goetzl EJ. (2001) Allergic diathesis in transgenic mice with constitutive T cell expression of inducible vasoactive intestinal peptide receptor. FASEB J, 15 (13): 2489-96. [PMID:11689474]

140. Voice JK, Grinninger C, Kong Y, Bangale Y, Paul S, Goetzl EJ. (2003) Roles of vasoactive intestinal peptide (VIP) in the expression of different immune phenotypes by wild-type mice and T cell-targeted type II VIP receptor transgenic mice. J Immunol, 170 (1): 308-14. [PMID:12496414]

141. Waschek JA, Casillas RA, Nguyen TB, DiCicco-Bloom EM, Carpenter EM, Rodriguez WI. (1998) Neural tube expression of pituitary adenylate cyclase-activating peptide (PACAP) and receptor: potential role in patterning and neurogenesis. Proc Natl Acad Sci USA, 95 (16): 9602-7. [PMID:9689127]

142. Wei Y, Mojsov S. (1996) Tissue specific expression of different human receptor types for pituitary adenylate cyclase activating polypeptide and vasoactive intestinal polypeptide: implications for their role in human physiology. J Neuroendocrinol, 8 (11): 811-7. [PMID:8933357]

143. Xia M, Sreedharan SP, Bolin DR, Gaufo GO, Goetzl EJ. (1997) Novel cyclic peptide agonist of high potency and selectivity for the type II vasoactive intestinal peptide receptor. J Pharmacol Exp Ther, 281 (2): 629-33. [PMID:9152366]

144. Yada T, Sakurada M, Ihida K, Nakata M, Murata F, Arimura A, Kikuchi M. (1994) Pituitary adenylate cyclase activating polypeptide is an extraordinarily potent intra-pancreatic regulator of insulin secretion from islet beta-cells. J Biol Chem, 269: 1290-1293. [PMID:8288592]

145. Yamamoto K, Hashimoto H, Tomimoto S, Shintani N, Miyazaki J, Tashiro F, Aihara H, Nammo T, Li M, Yamagata K et al.. (2003) Overexpression of PACAP in transgenic mouse pancreatic beta-cells enhances insulin secretion and ameliorates streptozotocin-induced diabetes. Diabetes, 52 (5): 1155-62. [PMID:12716746]

146. Yiangou Y, Di Marzo V, Spokes RA, Panico M, Morris HR, Bloom SR. (1987) Isolation, characterization, and pharmacological actions of peptide histidine valine 42, a novel prepro-vasoactive intestinal peptide-derived peptide. J Biol Chem, 262: 14010-14013. [PMID:3654650]

147. Zaben M, Sheward WJ, Shtaya A, Abbosh C, Harmar AJ, Pringle AK, Gray WP. (2009) The neurotransmitter VIP expands the pool of symmetrically dividing postnatal dentate gyrus precursors via VPAC2 receptors or directs them toward a neuronal fate via VPAC1 receptors. Stem Cells, 27 (10): 2539-51. [PMID:19650041]

148. Zhou CJ, Shioda S, Shibanuma M, Nakajo S, Funahashi H, Nakai Y, Arimura A, Kikuyama S. (1999) Pituitary adenylate cyclase-activating polypeptide receptors during development: expression in the rat embryo at primitive streak stage. Neuroscience, 93 (1): 375-91. [PMID:10430501]

149. Zupan V, Hill JM, Brenneman DE, Gozes I, Fridkin M, Robberecht P, Evrard P, Gressens P. (1998) Involvement of pituitary adenylate cyclase-activating polypeptide II vasoactive intestinal peptide 2 receptor in mouse neocortical astrocytogenesis. J Neurochem, 70 (5): 2165-73. [PMID:9572304]

How to cite this page

To cite this family introduction, please use the following:

Database page citation (select format):