Elsevier

Peptides

Volume 32, Issue 8, August 2011, Pages 1647-1655
Peptides

PAC1hop, null and hip receptors mediate differential signaling through cyclic AMP and calcium leading to splice variant-specific gene induction in neural cells

https://doi.org/10.1016/j.peptides.2011.06.004Get rights and content

Abstract

Pituitary adenylate cyclase-activating polypeptide (PACAP)-mediated activation of its G protein-coupled receptor PAC1 results in activation of the two G proteins Gs and Gq to alter second messenger generation and gene transcription in the nervous system, important for homeostatic responses to stress and injury. Heterologous expression of the three major splice variants of the rat PAC1 receptor, PAC1hop, null and hip, in neural NG108-15 cells conferred PACAP-mediated intracellular cAMP generation, while elevation of [Ca2+]i occurred only in PAC1hop-, and to a lesser extent in PAC1null-expressing cells. Induction of vasoactive intestinal polypeptide (VIP) and stanniocalcin 1 (STC1), two genes potentially involved in PACAP's homeostatic responses, was examined as a function of the expressed PAC1 variant. VIP induction was greatest in PAC1hop-expressing cells, suggesting that a maximal transcriptional response requires combinatorial signaling through both cAMP and Ca2+. STC1 induction was similar for all three receptor splice variants and was mimicked by the adenylate cyclase activator forskolin, indicating that cAMP elevation is sufficient to induce STC1. The degree of activation of two different second messenger pathways appears to determine the transcriptional response, suggesting that cellular responses to stressors are fine-tuned through differential receptor isoform expression. Signaling to the VIP gene proceeded through cAMP and protein kinase A (PKA) in these cells, independently of the MAP kinase ERK1/2. STC1 gene induction by PACAP was dependent on cAMP and ERK1/2, independently of PKA. Differential gene induction via different cAMP dependent signaling pathways potentially provides further targets for the design of treatments for stress-associated disorders.

Highlights

► PAC1hop, null and hip confer cAMP generation upon PACAP treatment in NG108-15 cells. ► PAC1hop and null confer elevation of [Ca2+]i, PAC1hop more efficaciously. ► Gene induction proceeds through PAC1hop-specific combinatorial or cAMP signaling. ► Gene induction through cAMP is mediated by PKA-dependent or -independent signaling.

Introduction

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a peptide that was isolated from ovine hypothalamus based on its ability to stimulate cyclic adenosine 3′5′-monophosphate (cAMP) in rat anterior pituitary cells [33]. The mature peptide PACAP occurs in two C-terminally α-amidated forms, PACAP-27 and PACAP-38, with PACAP-27 being identical to the first 27 amino acids of PACAP-38 [34]. PACAP-27 has 68% sequence homology with vasoactive intestinal polypeptide (VIP), identifying PACAP as a member of the VIP-secretin-growth hormone releasing hormone-glucagon superfamily. The first 27 amino acids of PACAP have almost been completely preserved through vertebrate evolution, from fish to mammals, and are responsible for its biological activity [37], [50]. PACAP is widely expressed in the nervous system, e.g., the hypothalamus, cerebral cortex, amygdala, nucleus accumbens, hippocampus and cerebellum of the central nervous system, and in sensory neurons, sympathetic preganglionic neurons and parasympathetic pre- and postganglionic neurons of the peripheral nervous system. PACAP-38 is the predominant form expressed [4], [18], [20], [56].

PACAP binds to three G protein-coupled receptors (GPCRs), named VPAC1, VPAC2 and PAC1, which are members of the class B family of GPCRs. VPAC1 and VPAC2 bind VIP and PACAP with similar affinity, whereas PAC1 binds PACAP with high affinity and VIP with much lower affinity. PACAP receptors are abundantly expressed in the central nervous system, the anterior pituitary and adrenal gland [2], [23], [24], [28], [31], [45], [52], [53]. Several isoforms of the PACAP-preferring PAC1 receptor have been identified in vivo. These are generated through alternative splicing within two regions of the PAC1 gene: the N-terminus and the third intracellular loop (ic3). N-terminal variants result from deletions (21 or 57 amino acids) at the N-terminal extracellular domain affecting ligand binding and the relative potencies of the ligands in second messenger stimulation [9], [43]. Ic3 variants result from the presence or absence of different insertions at the C-terminal end of the loop, a domain thought to be crucial for interaction with G proteins. Each insertion, designated hip and hop, consists of an 84-bp cassette. The alternative use of two contiguous consensus splice acceptor sites at the 5′-end of the hop cassette generates hop1 and hop2. The hip cassette can be included together with the hop cassette to give rise to hiphop. The null form does not contain any insert. The hop cassette encodes a consensus motif for phosphorylation by protein kinase C (PKC) [54]. In the adrenal medulla the predominant PAC1 variant is PAC1hop, in the brain PAC1hop and null are abundantly expressed. PAC1 with a full-length N-terminus is the predominant form in the adult brain, whereas the embryonic brain expresses high levels of receptors containing a short N-terminus lacking 21 amino acids [15], [27], [38], [40], [46], [68].

All PACAP receptors regulate cAMP generation by coupling to adenylate cyclases (ACs) through Gs. Coupling to phospholipase Cβ (PLCβ), in contrast, varies among the different receptor sub-types. Regulation of inositol phosphate (IP) production by coupling to PLCβ through Gq is more efficacious in PAC1hop and null compared to hip receptors as assessed in non-neural heterologous cells [27], [46], [54]. The functional importance of Gs/AC- and Gq/PLCβ-mediated combinatorial signaling has been shown for sustained release of catecholamines (CAs) and neuropeptides from adrenomedullary chromaffin cells (CCs) [5], [16], [29], [49], [63], which predominantly express the PAC1hop receptor variant [38], [40]. In the adrenomedullary pheochromocytoma PC12 cell line [14] it has been shown that PAC1hop-activated sustained CA release proceeds through inositol-1,4,5-trisphosphate (IP3)-mediated Ca2+ release from intracellular stores and store-operated Ca2+ entry (SOCE) [39], [58]. Moreover, the activation of two second messenger pathways, cAMP and Ca2+, seems to be required for maximal transcriptional stimulation of the neuropeptide VIP in CCs [17]. Stanniocalcin 1 (STC1) is another PACAP-regulated gene in CCs [1] with potentially neurotrophic functions [65], [66], [67]; signaling pathways regulating its neural expression, however, remain unidentified.

Although the different ic3 splice variants of PAC1 were first discovered almost 20 years ago, an understanding of second messenger production and gene induction mediated by these different variants in neural cells is still lacking. Therefore, we investigated the induction of the second messengers cAMP and Ca2+ as well as the PACAP target genes VIP and STC1 by the rat PAC1hop1, null and hip receptor variants with a full-length N-terminus in neural NG108-15 cells. The NG108-15 cell line is a neuroblastoma × glioma hybrid, not responding to PACAP endogenously, therefore providing an appropriate model system to study the different PAC1 splice variants separately introduced into a neural cell line. We demonstrate here that combinatorial signaling through cAMP and Ca2+, mediated uniquely by PAC1hop, is required for a full transcriptional response of the VIP gene. Cyclic AMP generation by either PAC1hop, null or hip is sufficient for induction of the gene encoding the neuroprotective protein STC1. Furthermore, two separate cAMP-dependent signaling pathways, activated by PACAP through PAC1hop, differentially regulate neural target genes. Our results provide evidence for the importance of differential expression of PAC1 splice variants and induction of second messenger pathways in shaping the PACAP-mediated transcriptional response in the nervous system.

Section snippets

Materials

PACAP-38 was purchased from Phoenix Pharmaceuticals (Mountain View, CA). Forskolin, H89, U0126 and 2′5′-dideoxyadenosine were obtained from Calbiochem (San Diego, CA). All cell culture media and supplements were obtained from Invitrogen (Carlsbad, CA) unless otherwise specified.

Culture of NG108-15 cells

NG108-15 cells (mouse neuroblastoma × rat glioma hybrid), obtained from the American Type Culture Collection (Manassas, VA) were cultured in high glucose Dulbecco's modified Eagle's medium (DMEM) supplemented with 10%

Generation of NG108-15 cells stably expressing the PAC1null, hop1 or hip receptor variant

To characterize each individual PAC1 splice variant separately in neural cells, the neuroblastoma × glioma cell line NG108-15 was transduced with the rat PAC1null, hop1 or hip receptor variants. NG108-15 cells expressed negligible levels of the PACAP receptors VPAC2 and PAC1hop endogenously (not shown) and a PACAP response could only occur via activation of reconstituted but not endogenous receptors. Thus, although NG108-15 cells are capable of G protein signaling through activation of endogenous

Discussion

PACAP is an important neuropeptide slow transmitter acting as a neurotrophic factor during brain development, a neuroprotective factor after brain injury and a regulator of the adrenal gland during prolonged stress [3], [37], [42], [51]. Its effects on prolonged secretion of epinephrine from the adrenal medulla and corticosterone from the adrenal cortex are accompanied by transcriptional changes that occur at the level of the adrenal gland but also the hypothalamus, reflecting the importance of

Disclosure statement

The authors have nothing to disclose.

Acknowledgments

We thank Dr. Laurent Journot for sharing their pRK8-PAC1 vectors, James Walsh for assistance in the subcloning of the PAC1 receptor variants, James Nagle and Debbie Kauffman (DNA Sequencing Facility, NINDS, National Institutes of Health) for carrying out sequencing of all DNA samples, Dr. Maribeth V. Eiden and members of her lab (NIMH, National Institutes of Health) for assistance in producing viral particles and Prof. E. Weihe (Philipps-University, Marburg, Germany) for advice and consultation

References (68)

  • A. Miyata et al.

    Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells

    Biochem Biophys Res Commun

    (1989)
  • A. Miyata et al.

    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

    (1990)
  • H. Morio et al.

    Pituitary adenylate cyclase-activating polypeptide protects rat-cultured cortical neurons from glutamate-induced cytotoxicity

    Brain Res

    (1996)
  • T. Mustafa et al.

    The hop cassette of the PAC1 receptor confers coupling to Ca2+ elevation required for pituitary adenylate cyclase-activating polypeptide-evoked neurosecretion

    J Biol Chem

    (2007)
  • T. Mustafa et al.

    PAC1hop receptor activation facilitates catecholamine secretion selectively through 2-APB-sensitive Ca(2+) channels in PC12 cells

    Cell Signal

    (2010)
  • C. Pantaloni et al.

    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

    (1996)
  • J.R. Pisegna et al.

    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

    (1996)
  • R.A. Rius et al.

    Pituitary adenylate cyclase activating polypeptide (PACAP) potently enhances tyrosine hydroxylase (TH) expression in adrenal chromaffin cells

    Life Sci

    (1994)
  • S. Shioda et al.

    Localization of the pituitary adenylate cyclase-activating polypeptide receptor and its mRNA in the rat adrenal medulla

    Neurosci Lett

    (2000)
  • S. Shioda et al.

    Localization and gene expression of the receptor for pituitary adenylate cyclase-activating polypeptide in the rat brain

    Neurosci Res

    (1997)
  • N. Stroth et al.

    Stress hormone synthesis in mouse hypothalamus and adrenal gland triggered by restraint is dependent on pituitary adenylate cyclase-activating polypeptide signaling

    Neuroscience

    (2010)
  • J. Tanaka et al.

    Neuronal protection from apoptosis by pituitary adenylate cyclase-activating polypeptide

    Regul Pept

    (1997)
  • T. Watanabe et al.

    Pituitary adenylate cyclase activating polypeptide provokes cultured rat chromaffin cells to secrete adrenaline

    Biochem Biophys Res Commun

    (1992)
  • K.Z. Zhang et al.

    High expression of stanniocalcin in differentiated brain neurons

    Am J Pathol

    (1998)
  • C.J. Zhou et al.

    Cellular distribution of the splice variants of the receptor for pituitary adenylate cyclase-activating polypeptide (PAC(1)-R) in the rat brain by in situ RT-PCR

    Brain Res Mol Brain Res

    (2000)
  • D. Ait-Ali et al.

    Neuropeptides, growth factors, and cytokines: A cohort of informational molecules whose expression is up-regulated by the stress-associated slow transmitter PACAP in chromaffin cells

    Cell Mol Neurobiol

    (2010)
  • A. Arimura

    Perspectives on pituitary adenylate cyclase activating polypeptide (PACAP) in the neuroendocrine, endocrine, and nervous systems

    Jpn J Physiol

    (1998)
  • A. Arimura et al.

    Tissue distribution of PACAP as determined by RIA: highly abundant in the rat brain and testes

    Endocrinology

    (1991)
  • S. Cavallaro et al.

    Pituitary adenylate cyclase activating polypeptide prevents apoptosis in cultured cerebellar granule neurons

    Mol Pharmacol

    (1996)
  • S. Cavallaro et al.

    Differentiation induces pituitary adenylate cyclase-activating polypeptide receptor expression in PC-12 cells

    Mol Pharmacol

    (1995)
  • F.M. Dautzenberg et al.

    N-terminal splice variants of the type I PACAP receptor: isolation, characterization and ligand binding/selectivity determinants

    J Neuroendocrinol

    (1999)
  • L.E. Eiden et al.

    Discovery of pituitary adenylate cyclase-activating polypeptide-regulated genes through microarray analyses in cell culture and in vivo

    Ann N Y Acad Sci

    (2008)
  • D. Frechilla et al.

    BDNF mediates the neuroprotective effect of PACAP-38 on rat cortical neurons

    Neuroreport

    (2001)
  • R.S. Ghadessy et al.

    Second messenger-dependent protein kinases and protein synthesis regulate endogenous secretin receptor responsiveness

    Br J Pharmacol

    (2002)
  • Cited by (34)

    • Signaling pathways and promoter regions that mediate pituitary adenylate cyclase activating polypeptide (PACAP) self-regulation in gonadotrophs

      2020, Molecular and Cellular Endocrinology
      Citation Excerpt :

      Moreover, the Hop-1 form was more effective than the Short form as an activator of PACAP transcription in LβT2 cells even though the level of receptor expression, both endogenously and with overexpression (Fig. 3A), was similar, implying variable activation of different signaling pathways. PAC1R is known to activate several signaling pathways including the PKA, PKC and MAPK pathways (May et al., 2010; Holighaus et al., 2011; Dickson and Finlayson, 2009), and while all isoforms stimulate cAMP production, the Hop1 isoform also stimulates PLD (McCulloch et al., 2000). BIM, H-89 and PD98059 were used to disrupt the PKC, PKA, and MAPK pathways, respectively, in un-stimulated and PACAP-stimulated αT3-1 and LβT2 cells.

    • PAC1 deficiency attenuates progression of atherosclerosis in ApoE deficient mice under cholesterol-enriched diet

      2020, Immunobiology
      Citation Excerpt :

      The PAC1 receptor interacts with PACAP exclusively, whereas VPAC1 and VPAC2 show comparable binding affinities to both PACAP and vasoactive intestinal polypeptide (VIP) (Ishihara et al., 1992; Lutz et al., 1993). All three receptors, upon activation, cause elevation of intracellular cyclic adenosine monophosphate (cAMP) via coupling to adenylyl cyclase, affecting proliferation, differentiation, remodeling and survival in various cell types (Holighaus et al., 2011; May and Parsons, 2017). They have been reported to be expressed in macrophages (MФ) (Delgado et al., 1999; Ganea and Delgado, 2001; Waschek, 2013), consistent with reported immunomodulatory effects of PACAP (Abad and Tan, 2018).

    • Pleiotropic pituitary adenylate cyclase-activating polypeptide (PACAP): Novel insights into the role of PACAP in eating and drug intake

      2020, Brain Research
      Citation Excerpt :

      The PAC1 receptor has several variants that result from alternative splicing of the PAC1 gene (Ushiyama et al., 2007). In the rat, most of these differences occur in the third intracellular loop, and are characterized by the absence (null variant) or presence of either one or two different insertions at the C-terminal end of the loop, which are 28 amino acid cassettes (hip or hop1 variant) or a 27 amino acid cassette (hop2 variant), that can be included together to form hiphop (Holighaus et al., 2011; Vaudry et al., 2009). While all PAC1 receptor variants regulate the production of cAMP, the null and hop1 variants have also been found to regulate phospholipase C production (Holighaus et al., 2011), while the presence of the hip cassette has been found to impair stimulation of cAMP and abolish stimulation of phospholipase C (Zhou et al., 2000).

    • Therapeutic potential of PACAP in alcohol toxicity

      2019, Neurochemistry International
    View all citing articles on Scopus
    View full text