Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 13.
Published in final edited form as: Wiley Interdiscip Rev Membr Transp Signal. 2012 Aug 9;1(6):789–803. doi: 10.1002/wmts.62

Coupling of P2Y receptors to G proteins and other signaling pathways

Laurie Erb 1,*, Gary A Weisman 1
PMCID: PMC4358762  NIHMSID: NIHMS668850  PMID: 25774333

Abstract

P2Y receptors are G protein-coupled receptors (GPCRs) that are activated by adenine and uridine nucleotides and nucleotide sugars. There are eight subtypes of P2Y receptors (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14), which activate intracellular signaling cascades to regulate a variety of cellular processes, including proliferation, differentiation, phagocytosis, secretion, nociception, cell adhesion, and cell migration. These signaling cascades operate mainly by the sequential activation or deactivation of heterotrimeric and monomeric G proteins, phospholipases, adenylyl and guanylyl cyclases, protein kinases, and phosphodiesterases. In addition, there are numerous ion channels, cell adhesion molecules, and receptor tyrosine kinases that are modulated by P2Y receptors and operate to transmit an extracellular signal to an intracellular response.

Introduction

P2Y receptors belong to the superfamily of heptahelical receptors found exclusively in eukaryotes that operate by binding or sensing signals outside the cell and then activating intracellular processes by coupling to heterotrimeric G proteins. The main signaling molecules or agonists recognized by the eight subtypes of P2Y receptors are shown in Table 1 and consist of adenine and uridine nucleotides (ATP, ADP, UTP, and UDP) and nucleotide sugars (UDP-glucose). These nucleotides and nucleotide sugars are released basally from many cell types and release is intensified in response to mechanical stress, oxygen deprivation, viral infection, or apoptotic stimuli through opening of pannexin 1 channels.14 In addition, ATP is stored in specialized secretory vesicles and released by exocytosis from nerve terminals, mast cells, chromaffin cells, and platelets. Platelets, in particular, have been shown to store ADP, ATP, UTP, and other factors in secretory vesicles called dense granules and release these stores upon platelet aggregation.5 The extracellular concentration of nucleotides is controlled by a family of ectoenzymes that catalyze the breakdown of nucleotides to nucleosides and by nucleoside transporters that transport nucleosides back into cells.6,7 Besides the P2Y receptors, two other families of nucleoside/nucleotide receptors have been identified in eukaryotes: the P1 receptors (A1, A2A, A2B, and A3), which are G protein-coupled receptors activated by adenosine, and the P2X receptors (P2X1, P2X2, P2X3, P2X4, P2X5, P2X6, and P2X7), which are ligand-gated ion channels activated by ATP. These receptors (reviewed in Refs 8 and 9), are expressed throughout the body and have important roles in heart and immune functions and neurotransmission.8,9

Table 1. P2Y Receptor Function and Coupling to G proteins.

Receptor Agonist G protein—Main Effector(s) Function
P2Y1 ADP Gq—PLCβ, Rac, Rho activation Platelet shape change/aggregation
Bone resorption
Leptin secretion from adipocytes
Mechanical and thermal nociception
Atherosclerosis
Angiogenesis
Synaptic plasticity
P2Y2 ATP/UTP Gq—PLCβ activation
Go—PLCβ, Rac activation
G12—Rho activation
Inhibition of bone formation and mineralization
Immune cell recruitment and phagocytosis
Vascular tone, inflammation, thrombosis
Blood pressure regulation
Intraocular pressure regulation
Mechanical and thermal nociception
HIV infection of T cells
Epithelial K+/Cl secretion
Pancreatic and renal functions
Liver regeneration
Wound healing
P2Y4 UTP Gq—PLCβ activation
Go—PLCβ activation
Visual and auditory transmission
Intestinal K+/Cl secretion
P2Y6 UDP Gq—PLCβ activation
G12/13—Rho activation
Bone resorption
Vascular tone and inflammation
Cardiac fibrosis
Macrophage cytokine/chemokine release
Microglial phagocytosis
Epithelial Cl secretion
P2Y11 ATP Gq—PLCβ activation
Gs—AC activation
Inhibition of neutrophil apoptosis
Negative regulation of TLR signaling
Pancreatic Cl secretion
Cell cycle arrest in endothelium
P2Y12 ADP Gi/o—AC inhibition
PLCβ, RhoA activation
Platelet aggregation
Microglial activation/migration
Dendritic cell activation/macropinocytosis
Peripheral anti-nociception, central nociception
P2Y13 ADP Gi/o—AC inhibition
PLCβ, RhoA activation
Bone formation
Liver uptake of HDL
Inhibition of ATP release from RBCs
Peripheral anti-nociception
P2Y14 UDP/UDP-glucose Gi/o—AC inhibition
PLCβ activation
Immune function/IL-8 release in epithelium
Gastric function/stomach contractility
Peripheral anti-nociception

Data derived from Refs 3,1167.

The general structure of P2Y receptors includes an extracellular N-terminus containing several potential N-linked glycosylation sites, seven transmembrane spanning domains that assist in forming the ligand binding pocket, three extracellular loops, three intracellular loops that participate in G protein coupling, and an intracellular C-terminus that contains several consensus binding/phosphorylation sites for protein kinases.10 Studies with selective P2Y receptor agonists and antagonists or mice lacking specific P2Y receptor subtypes have demonstrated an array of physiological functions coupled to these receptors,3,1167 which are summarized in Table 1.

P2Y Receptor Coupling to G Proteins

Heterotrimeric G proteins consist of a Gα subunit that is tightly associated with Gβγ subunits and bind to G protein-coupled receptors (GPCRs) at the inner surface of the cell. P2Y receptors, like other members of the GPCR superfamily, act as guanine nucleotide exchange factors (GEFs) for heterotrimeric G proteins, causing dissociation of the G protein from the activated receptor as well as dissociation of the Gα subunit from the Gβγ complex. These separated G protein subunits can then interact with a variety of effector proteins, leading to either activation or deactivation of the effector protein. On the basis of the sequence similarity of the Gα subunits, heterotrimeric G proteins have been broadly divided into four subfamilies (Gq, Gs, Gi/o, and G12/13) that are used to define both receptor and effector coupling.43 Similarly, the P2Y receptors have been grouped into two subfamilies based on their sequence similarity: the Gq-coupled P2Y receptors, which share a 28–52% sequence homology and include the P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11 receptors, and the Gi/o-coupled P2Y receptors, which share a 45–50% sequence homology and include the P2Y12 P2Y13, and P2Y14 receptors. There are, however, anomalies to this classification in that most of the Gq-coupled P2Y receptors are known to link to other G protein subfamilies. These additional G protein linkages are shown in Table 1.

The ability of individual P2Y receptor subtypes to couple to specific G proteins was initially derived from indirect evidence obtained by measuring the cytoplasmic levels of second messengers, i.e., inositol phosphate, Ca2+ and cyclic AMP (cAMP), in response to nucleotide stimulation or by determining the sensitivity of nucleotide-induced signaling to the Gi/o protein inhibitor pertussis toxin (PTX). More recently, direct evidence for P2Y1 receptor coupling to Gq proteins (Gαqβ1γ2 and Gα11β1γ2) was obtained by measuring agonist-induced GTP hydrolysis in vesicles reconstituted with these proteins.44 Similarly, vesicle reconstitution of the P2Y12 receptor with specific G proteins indicates that this receptor couples to Gαi2 more effectively than to Gαi1 and Gαi3, but not to Gαo or Gαq.45 Coupling of the P2Y1 receptor to Gq results in the activation of PLCβ and mobilization of intracellular Ca2+ as well as the activation of the monomeric G proteins, RhoA and Rac,46 while coupling of the P2Y12 receptor to Gi2 results in the inhibition of adenylyl cyclase as well as the activation of PI3K, Akt, Rap1b, and potassium channels. Together, the P2Y1 and P2Y12 receptors, which are activated by ADP, control platelet shape change and aggregation and their effects have been extensively reviewed.35,47

The P2Y2 receptor is activated equally well by ATP or UTP and couples to Gq, Go, and G12 proteins, which was determined by measuring GTPγ [35S] binding to specific Gα subunits in response to agonist stimulation in 1321N1 astrocytoma cells.48,49 These studies also indicate that coupling of the P2Y2 receptor to Go and G12 requires interaction with αv integrins via a three amino acid integrin-binding domain (RGD) in the first extracellular loop of the P2Y2 receptor to access and activate Go and G12, whereas coupling to Gq does not require integrin interaction.4850 Activation of Go by the P2Y2 receptor leads to activation of RhoA, whereas activation of G12 leads to activation Rac and the RacGEF, Vav2, which together are important for cytoskeletal rearrangements involved in cell migration and phagocytosis (see Figure 1). By using antisense oligonucleotides against specific G protein subunits, the P2Y2 receptor in HEL cells was shown to activate PLCβ and mobilize intracellular Ca2+ by coupling to Gα16, a member of the Gq subfamily51 and to activate PLCβ1 and PLCβ3 in gastric smooth muscle cells via coupling to Gαq and the βγ subunits of Gαi3β1γ2, respectively.52

Figure 1.

Figure 1

Model of cytoskeletal signaling by the P2Y2 receptor through Go and G12 proteins. The P2Y2 receptor, which is linked to the actin cytoskeleton through association with filamin A,67 initiates cytoskeletal rearrangements involved in cell migration, neurite extension and phagocytosis by promoting the growth of actin filaments near the activated receptor. This is accomplished, in part, by the interaction of the P2Y2 receptor RGD domain with αVβ3/5 integrins.50 Integrin interaction allows the P2Y2 receptor to access and activate Go and G12 proteins that are known to associate with β3 integrins and with the integrin-associated protein, CD47.49,50 Activation of Go and G12 leads to activation of Rac1 and RhoA, which then activate the actin-binding LIM kinases (LIMKs) responsible for serine-phosphorylation/deactivation of the actin disassembly protein, cofilin.48,49

The P2Y4 receptor is activated by UTP and couples to Gq and Go proteins to activate PLCβ and mobilize intracellular Ca2+.53,54 This receptor also activates Rho and inhibits K+ currents in myocytes from rat cerebral arteries in a Rho-dependent manner.55

The P2Y6 receptor is activated by UDP and couples to members of the Gq and G12/13 subfamilies. In most cell types tested, the P2Y6 receptor couples to Gq to activate PLCβ and mobilize intracellular Ca2+, whereas coupling to Go appears to be absent.10 However, in rat sympathetic neurons injected with P2Y6 receptor mRNA, PTX inhibits the decrease in voltage-gated Ca2+ currents mediated by this receptor, suggesting that the P2Y6 receptor, in some instances, may operate through Gi/o.56 Coupling of the P2Y6 receptor to G12/13 and Rho activation was recently demonstrated to contribute to pressure overload-induced cardiac fibrosis in an in vivo mouse model.57 Nishida et al. generated mice with a cardiomyocyte-specific overexpression of p115-RGS, a regulator of the G protein signaling that specifically inhibits Gα12/13, to show that these mice have suppressed expression of fibrogenic genes in cardiomyocytes in response to mechanical stretch that is dependent on Rho activation. The authors also demonstrated that the mechanism of mechanical stretch-induced fibrosis involves the release of ATP and UDP from cardiomyocytes through pannexin 1 channels that activate the P2Y6 receptor.57

The P2Y11 receptor is activated by ATP and couples to members of the Gq and Gs subfamilies. Gs proteins stimulate adenylyl cyclase to increase production of cAMP and coupling of the P2Y11 receptor to Gs is important for controlling several physiological processes.36,58,59 In human monocytes, activation of the P2Y11 receptor negatively regulates toll-like receptor signaling by increasing cAMP production.58 Activation of P2Y11 receptors inhibits neutrophil apoptosis by a cAMP-dependent mechanism36 and activates cAMP-dependent Cl channels on the baso-lateral surface of pancreatic duct cells.59 Although ATP appears to be the main agonist for the P2Y11 receptor, UTP has been reported to stimulate Ca2+ mobilization in P2Y11 receptor-transfected 1321N1 astrocytoma cells by a PTX-sensitive mechanism, whereas Ca2+ mobilization induced by ATP in these cells is insensitive to PTX, suggesting that the P2Y11 receptor is capable, in some instances, of coupling to Go in addition to Gq.60 It is uncertain why the agonist profile and G protein coupling of the P2Y11 receptor is altered in this cell expression system, although it is possible that these differences may be due to the ability of the P2Y11 receptor to heterodimerize with other GPCRs. In support of this possibility, co-expression of the P2Y1 and P2Y11 receptors in HEK cells was found to alter the agonist profile from that in cells expressing only the P2Y11 receptor.61

The P2Y12 receptor is activated by ADP and couples mainly to Gi, although Go coupling is indicated in some cell types, as is PTX-insensitive signaling by the P2Y12 receptor.62 Soulet et al.62 have shown that expression of the P2Y12 receptor in CHO cells mediates PTX-sensitive cell proliferation that relies on signaling through PI3-kinase/Akt and ERK1/2, and PTX-insensitive actin cytoskeleton organization that relies on signaling through RhoA/Rho-kinase. Conversely, in C6 glioma cells, activation of the P2Y12 receptor enhances cell proliferation by a pathway that is dependent on Gαi, RhoA/ROCK, and PKCζ, but does not involve ERK1/2 signaling.63 In mouse dendritic cells, where the P2Y12 receptor is endogenously expressed, agonist stimulation of the P2Y12 receptor causes a PTX-sensitive increase in cytoplasmic Ca2+ that is necessary for macropinocytosis of antigens,12 suggesting that the P2Y12 receptor in dendritic cells can couple to Go.

The P2Y13 receptor couples to Gi/o and is activated by ADP. In transfected 1321N1 or CHO cells, agonist-induced activation of the P2Y13 receptor increases the PTX-sensitive production of IP3 and inhibits forskolin-mediated cAMP production, whereas high concentrations of P2Y13 receptor agonists cause an increase in cAMP production, suggesting that the P2Y13 receptor can, in some instances, couple to Gs.64 In human hepatocytes, endocytosis of HDL mediated by the P2Y13 receptor requires the activation of RhoA and ROCK I, although the subtype of G protein involved in this process was not confirmed.65 Similarly, the P2Y13 receptor activates RhoA and ROCK I in mouse osteoblasts and plays an important role in bone formation.17 In red blood cells, Wang et al.40 demonstrate that activation of the P2Y13 receptor decreases the release of ATP by these cells, by inhibiting the production of cAMP.

The P2Y14 receptor couples to Gi/o and is activated by UDP, UDP-glucose, and other UDP-linked sugars, including UDP-galactose and UDP-N-acetylglucosamine. The signaling properties of this receptor were recently reviewed66 and its physiological functions are beginning to be revealed. Studies in mouse and human tissues indicate that the P2Y14 receptor is highly expressed in mucosal epithelium where its activation leads to the production of chemokines and cytokines involved in the recruitment of neutrophils, including IL-8, macrophage inflammatory protein-2, and keratinocyte-derived cytokine.38 Also, high levels of the P2Y14 receptor are expressed in the rat forestomach where it plays a role in muscle contractility.13

Other Mechanisms of P2Y Receptor Signaling

P2Y Receptor Oligomerization

It is generally accepted that GPCRs can dimerize to form heteromeric and homomeric structures or more complex oligomeric structures and, accordingly, several examples of dimerization involving P2Y receptors have been characterized by performing co-immunoprecipitation experiments and/or FRET analysis. Both the P2Y1 and P2Y2 receptors form a heteromeric complex with the A1 adenosine receptor in HEK293 cells.68,69 Heteromeric association between the Gi-coupled A1 receptor and the Gq-coupled P2Y1 receptor was shown to create a hybrid receptor that responds predominantly to the P2Y1 receptor agonist ADPβS to activate both Gi and Gq.69 Specifically, activation of Gi and inhibition of forskolin-induced cAMP accumulation by the A1-P2Y1 receptor complex displays a hybrid selectivity for P2Y1 and A1 receptor agonists and antagonists, whereas activation of Gq and generation of inositol phosphates occurs only with P2Y1 receptor agonists and not with A1 receptor agonists,69 suggesting that the cross talk to G proteins for the A1–P2Y1 receptor complex is unidirectional. In contrast, association of the A1 receptor with the P2Y2 receptor does not affect the ligand selectivity of the heteromeric receptor, although simultaneous stimulation of the A1–P2Y2 receptor complex with A1 and P2Y2 receptor agonists interferes with Gi signaling and enhances Gq signaling.68

The P2Y1 receptor also is thought to form a heteromeric complex with the P2Y11 and P2Y12 receptor,61,70 which for the P2Y11 receptor alters its function. Ecke et al. have shown that co-expression of the P2Y1 and P2Y11 receptors in HEK293 or 1321N1 cells promotes agonist-induced endocytosis of the P2Y11 receptor, which when expressed alone in these cells is unable to undergo agonist-induced endocytosis. The ligand selectivity of the P2Y1–P2Y1161 receptor complex, measured by activation of Ca2+ signaling, is also different than the agonist potency profile in cells expressing only the P2Y11 receptor.

Recent publications show that the P2Y4, P2Y6, and P2Y12 receptors form homo-oligomeric complexes that are thought to be involved in receptor signaling.71,72 Importantly, Savi et al.71 have shown that clopidogrel, an antithrombotic agent that antagonizes the platelet P2Y12 receptor, interferes with homo-oligomeric assembly of the P2Y12 receptor and localization of these complexes into lipid rafts that is required for receptor activation. D'Ambrosi et al.72 have shown that in neuronal cells the P2Y4 and P2Y6 receptors form homo- and hetero-oligomeric complexes, that UTP affects the oligomerization of the P2Y6 receptor, but not the P2Y4 receptor, and that dimeric P2Y4 receptors and monomeric P2Y6 receptors selectively partition into lipid rafts from specialized subcellular compartments, such as synaptosomes.

P2Y Receptor Desensitization and Trafficking

Agonist-induced activation of GPCRs often initiates receptor desensitization, which diminishes GPCR responsiveness to ligands and can lead to receptor internalization or endocytosis. In addition, the kinetics of GPCR desensitization and recovery from desensitization was recently demonstrated to be required for sufficient activation of store-operated, calcium release-activated calcium (CRAC) channels that are tightly linked to expression of the c-fos transcripition factor, indicating that GPCR desensitization is also important for activating gene expression.73 Agonist-dependent GPCR desensitization is generally mediated by a family of GPCR kinases (GRK 1-7), which phosphorylate serine/threonine residues in intracellular domains of the receptor and, in some instances, promote the binding of β-arrestins (β-arrestin1-4). β-arrestins, in turn, can regulate several receptor functions, including desensitization (by inhibiting GPCR/G-protein interaction), endocytosis (by interacting with clathrin and AP2 to promote receptor internalization into clathrin-coated pits) and signaling (by interacting with MAPKs and Src).74 A recent study looked at P2Y1,2,4,6,11,12 receptor interactions with β-arrestins in transfected HEK-293 cells using FRET analysis or by measuring β-arrestin translocation to the plasma membrane in response to receptor activation.75 A summary of these interactions is shown in Table 2. In this study, Hoffmann et al. determined that all six of these P2Y receptor subtypes interact to some degree with β-arrestin2, whereas the P2Y2 and P2Y4 receptors interact with both β-arrestin1 and β-arrestin2. Interestingly, they found that the P2Y2 receptor couples differently to β-arrestins depending on whether ATP or UTP is used to activate this receptor; activation of the P2Y2 receptor with ATP causes a strong interaction with β-arrestin1 and a weak interaction with β-arrestin2, whereas activation of the P2Y2 receptor with UTP causes a strong interaction with both β-arrestin1 and β-arrestin2. This differential coupling of the P2Y2 receptor to β-arrestins in response to stimulation with ATP or UTP supports earlier findings indicating that desensitization of the P2Y2 receptor requires 10-fold higher concentrations of ATP than UTP, even though these nucleotides activate P2Y2 receptor signaling processes with similar potencies.76

Table 2. P2Y Receptor Trafficking.

Receptor Coupling to β-Arrestins Mechanism of β-Arrestin Coupling Mechanism of Internalization Apical/Basolateral Expression in Polarized Cells Mechanism of Coupling to the Cytoskeleton
P2Y1 β-arrestin2 PKC phosphorylation Dynamin Basolateral (dep. on basic/acidic aa ratio in the C-terminus)
P2Y2 β-arrestin2 (ATP)
β-arrestin1 and 2 (UTP)
GRK2 phosphorylation Dynamin-independent Apical (RGD domain-dep.) Filamin A
P2Y4 β-arrestin1 and 2 GRK phosphorylation Dynamin Apical
P2Y6 β-arrestin2 GRK2 phosphorylation Dynamin Apical
P2Y11 β-arrestin2 GRK2 phosphorylation Dynamin, dimerization with P2Y1 Basolateral
P2Y12 β-arrestin2 GRK2,6 phosphorylation Dynamin, PDZ domain-dep. Basolateral
P2Y13 unknown Unsorted
P2Y14 unknown Basolateral

Data derived from Refs 67,75,7785.

β-arrestins are, in part, responsible for GPCR-mediated activation of the MAPKs, ERK1/2, by inducing receptor endocytosis and relocalization to endosomes.74 The P2Y2 receptor, which displays differential coupling to β-arrestins in response to activation by ATP versus UTP, also displays a differential effect on the duration of ERK1/2 activation in response to these endogenous P2Y2 receptor ligands (i.e., ATP causes sustained ERK1/2 activation, whereas UTP causes transient ERK1/2 activation).75 In numerous studies, the duration of ERK1/2 activation is critical for determining cell fate (i.e., whether cells differentiate, proliferate, procreate or die).86 This suggests that the P2Y2 receptor may regulate different cell fates, depending on which receptor agonist is present.

In rat mesenteric smooth muscle cells, β-arrestin2 and GRK2 are required for P2Y2 receptor desensitization and termination of p38 and ERK1/2 signaling.77,78 Receptor mutagenesis studies indicate that deletion of structural motifs or point mutations of putative GRK phosphorylation sites (e.g., S243A, T344A, and S356A) in the P2Y2 receptor C-terminal tail diminishes agonist-induced desensitization and internalization of this receptor.79,80 Suppression of specific GRKs with siRNA demonstrate that desensitization of the P2Y12 receptor requires GRK2 and GRK6 activity.81 In contrast, P2Y1 receptor desensitization is largely dependent on PKC activity.81 In HEK293 cells, a dominant negative dynamin construct (dyn-K44A) was used to demonstrate that agonist-induced internalization of the P2Y1,4,6,11,12 receptors is dynamin-dependent, whereas agonist-induced internalization of the P2Y2 receptor is dynamin-independent.75

Activation of receptor tyrosine kinases by growth factors has been shown to resensitize P2Y receptors in many different cell types by PKC-dependent P2Y receptor phosphorylation.87,88 In retinal glial cells, P2Y receptors are desensitized by ATP and resensitized by growth factors (e.g., EGF, PDGF, NGF), via activation of PI3K and protein phosphatases, whereas resensitization is not dependent on the activity of PKC, Src kinases, or ERK1/2.89

Many GPCRs express C-terminal PDZ-binding domains (type I: X–S/T–X–O, type II: X–O–X–O, type III: X–D/E–X–O, where X = any and O = hydrophobic amino acid) that interact with PDZ domain-containing proteins and have established roles in receptor trafficking, localization, and assembly of signaling complexes.90 Several P2Y receptors contain PDZ-binding domains, including the P2Y1,2,4,12,14 receptors, and the PDZ-binding domain in the extreme C-terminal tail of the P2Y12 receptor (ETPM) has been shown to be important for receptor trafficking in platelets.82 Disruption or removal of the P2Y12 PDZ-binding domain blocks interaction with β-arrestin and slows receptor internalization in CHO cells, which the authors suggest is either due to interaction of the PDZ-binding domain with a regulatory kinase or phosphorylation of the threonine residue within the PDZ-binding domain that is required for β-arrestin interaction.82

Polarized expression of P2Y receptors in apical versus basolateral surfaces of epithelial cells has been observed (see Table 2). Wolff et al.83 demonstrated that the P2Y1,11,12,14 receptors are basolaterally expressed, whereas the P2Y2,4,6 receptors are apically expressed and the P2Y13 receptor is unsorted in polarized Madin-Darby canine kidney (MDCK) cells. Mutagenesis studies with the P2Y1 and P2Y2 receptors indicate that charged amino acid residues (five basic and four acidic) in the C-terminal tail of the P2Y1 receptor are important for basolateral trafficking,84 whereas the RGD domain in the first extracellular loop of the P2Y2 receptor is important for apical trafficking in polarized MDCK cells.85

P2Y Receptor Cross Talk

Receptor cross talk refers to the simultaneous activation of different types of cell surface receptors resulting in coordinated stimulation or suppression of signal transduction. Cross talk with P2Y receptors can produce a synergistic or more than additive change in the level of second messengers. For example, co-activation of various Gi-coupled receptors and Gq-coupled P2Y receptors induces a synergistic rise in intracellular IP3 and Ca2+ levels9194 and the release of arachidonic acid (AA)93,95, as compared to activation of Gq-coupled P2Y receptors alone. Also, prestimulation of Gq-coupled P2Y1 or P2Y2 receptors in HEK293 cells enables the Gi-coupled CXCR2 chemokine receptor and the Gs-coupled β–adrenergic receptor to mobilize intracellular Ca2+, a response not normally regulated by these Gi- and Gs-coupled receptors.94 Gq-coupled P2Y receptors stimulate PLC activity that generates DAG, an endogenous activator of PKC, which in turn can activate phospholipase A2 and generate AA from membrane phospholipids. Although activation of Gi-coupled m2 muscarinic, α2 adrenergic, and D2 dopaminergic receptors in CHO-K1 cells does not induce AA release, these receptor activities can enhance the effect of P2Y receptor activation on AA generation.95 Activation of the P2Y2 receptor resensitizes vanilloid type1 channels (TRPV1) in kidney sensory neurons96 and activation of the P2Y1 receptor causes PKC-dependent phosphorylation of the capsaicin receptor (a VR1 cation channel), which shifts the capsaicin concentration–response curve twofold to the left, and decreases the threshold for capsaicin receptor activation by heat from 42 to 35°C.97 Thus, cross talk between the P2Y1 and capsaicin receptors may represent a novel mechanism for the perception of pain (nociception) induced by P2Y receptor activation.

Cross talk between P2Y receptors and P2X receptor ion channels has also been demonstrated. In Xenopus oocytes expressing the recombinant human P2X1 receptor, a transient inward current occurs in response to ATP.98 This response undergoes desensitization (i.e., current flow decreases upon prolonged or repeated exposure to ATP), however, co-expression and co-activation of either the P2Y1 or P2Y2 receptor (stimulated by ADP or UTP, respectively) inhibits P2X1 receptor desensitization. The mechanism of P2Y receptor-mediated inhibition of P2X1 receptor desensitization does not appear to involve direct phosphorylation of the P2X1 receptor, but does involve protein kinase activity perhaps mediated by an accessory protein.98 P2X1 receptors can also modulate P2Y receptor activity. In megakaryocytes, P2X1 receptor activation with α,β-meATP causes a rapid and transient Ca2+ influx, whereas P2Y1 receptor activation with ADP causes a slower, larger, and more sustained increase in cytoplasmic Ca2+.99 Co-application of the two agonists, however, accelerates the Ca2+ response and potentiates the peak amplitude, suggesting that the P2X1 receptor may have a priming role in the activation of P2Y1 receptors during platelet stimulation. The P2Y12 receptor, through activation of PI3K, also has been shown to play a synergistic role in the regulation of both P2Y1 and P2X1 receptor-mediated currents in megakaryocytes100 and activation of P2Y2 receptors resensitizes P2X2 receptor channels in bladder sensory neurons.101

P2Y Receptor Coupling to Ion Channels

The generation of knock-out (KO) mouse models has helped to identify ion channels controlled by various subtypes of P2Y receptors, which have been extensively reviewed.10,102,103 In respiratory epithelium, the generation of the P2Y2 receptor KO mouse helped to show that the major luminal P2Y receptor involved in the activation of Cl secretion is the P2Y2 receptor subtype. Not all epithelia express the P2Y2 receptor subtype luminally, but still respond robustly to the addition of luminal UTP. Mouse jejunum displays luminal UTP-stimulated Cl secretion, which was not affected in P2Y2 receptor KO mice.104 Using a P2Y4 receptor KO mouse, it was recently established that the P2Y4 receptor is responsible for the UTP-stimulated Cl secretion.105

The P2Y1 receptor contains a PDZ-binding domain (DTSL) in its C-terminal tail that interacts directly with the Na+/H+-exchange regulatory factor (NHERF-2) to control Na+/H+ exchange.106 Similarly, an RRSE-QXK/RSE motif that is present in P2Y1,2,6,11 receptors is required for modulation of a voltage-gated ion channel that mediates a transient inward current in Xenopus oocytes.107

Activation of the P2Y2 receptor in many cell types causes the cytoplasmic calcium-dependent entry of extracellular calcium,108,109 which Dong et al.110 have shown to be caused by the opening of store-operated, CRAC channels. In airway epithelium, activation of the P2Y2 receptor increases Cl secretion by opening a calcium-activated chloride channel and the cystic fibrosis transmembrane conductance regulator (CFTR), which has potential relevance to the treatment of cystic fibrosis, a disease that is caused by mutations in the CFTR gene.111113 The synthetic P2Y2 receptor agonist INS37217 has been employed to promote Cl and water secretion in tracheal epithelium, to increase ciliary beat frequency and mucin release in human airway epithelium114 and to stimulate subretinal fluid reabsorption in a rabbit model of retinal detachment.115 Both the P2Y2 and P2Y4 receptors are present in the luminal membrane of mouse distal colonic mucosa and stimulation of these receptors increases K+ secretion.23

In the cochlea, activation of the P2Y4 receptor in epithelial cells of Reissner's membrane inhibits Na+ transport out of endolymph by closing the epithelial Na+ channel (ENaC), which the authors speculate might be caused by a decrease in the open probability of ENaC due to depletion of PI(4,5)P2 in the plasma membrane after P2Y4 receptor activation.116 The P2Y4 receptor also controls the lumenal K+ concentration in the cochlea by activating apical potassium channels on strial marginal epithelial cells117, whereas P2Y4 inhibits K+ currents in cerebral artery myocytes.55 P2Y1,2,6 receptors couple to neuronal N-type Ca2+ channels and to M-type K+ channels, whereas the P2Y4 receptor couples more effectively to M-type K+ channels than to Ca2+ channels and involves the βγ subunits of Gi/o proteins.118

Gi-coupled receptors, including the P2Y12,13,14 receptors, have been shown to inhibit nociception in peripheral neurons by blocking voltage-gated Ca2+ channels.119 Many P2Y receptors, including the P2Y1,2,4,6,12 subtypes, have been shown to modulate G protein-gated, inwardly rectifying K+ (GIRK) channels,103 which are known to mediate the antinociceptive actions of opioids in the spine.120 In general, opening of GIRK channels by P2Y receptors and other GPCRs is mediated by the βγ subunit of Gi/o proteins, whereas closing of GIRK channels is mediated by the α subunit of Gq proteins.103,121 The P2Y1 receptor, which couples primarily to Gq proteins, has been shown to both open and close GIRK channels when expressed and activated in rat sympathetic neurons or CHO cells.121,122 Furthermore, closing of GIRK channels mediated by the P2Y1 receptor in these studies was PTX-sensitive, indicating that the P2Y1 receptor in this instance is capable of coupling to Gi/o proteins. Another potassium channel implicated in pain perception, the two-pore K+ channel (K2P), is generally closed by GPCRs coupled to Gq or Gs and opened by GPCRs coupled to Gi/o.123 Shrestha et al., however, demonstrated that activation of both the Gq-coupled P2Y1 receptor and the Gi/o-coupled P2Y12 receptor inhibits K2P activity in tsA201 embryonic kidney cells, which the authors suggest is due to the formation of a P2Y1–P2Y12 receptor complex that suppresses Gi/o signaling,70 similar to the alteration in G protein signaling observed for the A1-P2Y2 receptor complex.68

P2Y Receptor Coupling to Receptor and Non-Receptor Tyrosine Kinases

Many studies show that P2Y receptors modulate the activity of growth factor receptors or receptor tyrosine kinases as well as non-receptor tyrosine kinases, such as Src.10 For example, studies with the P2Y2 receptor indicate that SH3-binding domains (PXXP) in the C-terminal tail are necessary for the constitutive association with the actin-binding protein, filamin A,67 as well as with the transient association and activation of Src.124 This, then, is necessary for Src-dependent activation of the focal adhesion-related kinase (RAFTK or Pyk2) and several receptor tyrosine kinases, including the platelet-derived growth factor receptor (PDGFR) and epidermal growth factor receptor (EGFR), by the P2Y2 receptor (see Figure 2).124 In endothelial cells, the P2Y2 receptor uses this Src-dependent mechanism to transactivate the vascular endothelial growth factor receptor-2 (VEGFR-2), leading to increased expression of VCAM-1, a vascular cell adhesion molecule that allows firm attachment of circulating leukocytes to the vascular wall.125 Recently, the P2Y1 receptor was shown to transactivate VEGFR-2 to stimulate endothelial cell tubulogenesis,30 indicating that multipe P2Y receptor subtypes can use VEGFR-2 signaling pathways to affect endothelial cell function.

Figure 2.

Figure 2

Model of P2Y2 receptor signaling to Gq proteins, ion channels, and growth factor receptors. Stimulation of the P2Y2 receptor with ATP or UTP causes Gαq-dependent activation of phospholipase C-β (PLCβ) resulting in the hydrolysis of PtdIns(4,5)P2 to produce the second messengers Ins(1,4,5)P3 (IP3), which mobilizes intracellular Ca2+, and diacylglycerol (DAG), which activates protein kinase C (PKC).10 Downstream signaling molecules include calmodulin (CaM), calmodulin-dependent kinase (CaMK), nitric oxide synthase (NOS), nitric oxide (NO), guanylyl cyclase (GC), cyclic GMP (cGMP), protein kinase G (PKG), cytosolic phospholipase A2 (cPLA2), arachidonic acid (AA), and prostaglandin E2 (PGE2). The P2Y2 receptor transactivates several growth factor receptors, including EGFRs, PDGFR, and VEGFR-2, through Src-dependent activation of the focal adhesion-related tyrosine kinase (Pyk2), which phosphorylates growth factor receptors and leads to activation of serine/threonine kinases (e.g., ERK1/2, JNK, p38, LIMK1, and p90RSK) and transcription factors (e.g., CREB, NF-kb, ELK-1, c-FOS, and c-JUN) that regulate genes involved in inflammation, apoptosis, cell differentiation, and so on.10 Src, which binds to Src homology 3 (SH3) domains in the P2Y2 receptor,124 also is involved in the activation of the monomeric G protein (Rac1) and p21-activated serine kinase (PAK) as well as phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt). The P2Y2 receptor also transactivates EGFRs by activating metalloproteases (ADAM10 and ADAM17), causing the cleavage and release of EGFR ligands, neuregulin and heparin-binding EGF (HB-EGF), which directly activate EGFRs.126,127

ATP is released upon epithelial injury and the P2Y2 receptor is known to be involved in epithelial wound repair by activating EGFRs.31,126,127 Studies have shown that the P2Y2 receptor in epithelium activates the metalloproteases ADAM10 and ADAM17, which catalyze the release of the growth factors heparin-binding EGF127 and neuregulin.126 These growth factors can then activate EGFRs in wounded epithelium to promote tissue repair.127

Van Kolan et al.128 demonstrated that activation of PKB by the P2Y12 receptor in C6 glioma cells requires the tyrosine kinase activities of the insulin growth factor-I (IGF-I) receptor, Src and Pyk2. Furthermore, in cells transfected with the Gβγ -scavenger β-ARK1/GRK2 or Rap1GAPII, stimulation of the P2Y12 receptor with 2-meSADP failed to enhance PKB phosphorylation demonstrating the requirement of Gβγ-subunits and Rap1 for P2Y12 receptor-mediated PKB activation. In CHO cells, activation of the P2Y12 receptor causes an increase in cell proliferation via pathways involving the activation of PI3K, ERK1/2, and the PDGF-β receptor.62

Conclusion

As described in this review, P2Y receptors for extracellular nucleotides have been shown to regulate cellular functions through the formation of multi-protein signaling complexes with other GPCRs, ion channels, integrins, growth factor receptors, cytoskeletal and junctional proteins, as well as with heterotrimeric and monomeric G proteins and other cytoplasmic signaling molecules. These interactions not only influence intracellular signaling induced by extracellular nucleotides, but also enable an individual P2Y receptor subtype to respond differently to nucleotides, depending on the repertoire of proteins expressed in a particular cell type. Since mammalian cells express 39 distinct G-protein subunits, i.e., 21 α-subunits, 6 β-subunits, and 12 γ-subunits, a myriad of heterotrimeric combinations likely exist that potentially regulate the functional consequences of P2Y receptor activation and the divergence of P2Y receptor-mediated responses between different tissue types. Similarly, tissue-specific expression of P2Y receptor subtypes adds to the complexity of signals transduced by nucleotides in different tissues. Thus, the signaling complexes in which P2Y receptors play a functional role can mediate a wide variety of physiological and pathological effects through protein:protein interactions that are now being revealed, many of which are discussed and diagrammed in this review.

References

  • 1.Chekeni FB, Elliott MR, Sandilos JK, Walk SF, Kinchen JM, Lazarowski ER, Armstrong AJ, Penuela S, Laird DW, Salvesen GS, et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature. 2010;467:863–867. doi: 10.1038/nature09413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bao L, Locovei S, Dahl G. Pannexin membrane channels are mechanosensitive conduits for ATP. FEBS Lett. 2004;572:65–68. doi: 10.1016/j.febslet.2004.07.009. [DOI] [PubMed] [Google Scholar]
  • 3.Seror C, Melki MT, Subra F, Raza SQ, Bras M, Saidi H, Nardacci R, Voisin L, Paoletti A, Law F, et al. Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. J Exp Med. 2011;208:1823–1834. doi: 10.1084/jem.20101805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bargiotas P, Krenz A, Hormuzdi SG, Ridder DA, Herb A, Barakat W, Penuela S, von Engelhardt J, Monyer H, Schwaninger M. Pannexins in ischemia-induced neurodegeneration. Proc Natl Acad Sci USA. 2011;108:20772–20777. doi: 10.1073/pnas.1018262108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lazarowski ER, Boucher RC. UTP as an extracellular signaling molecule. News Physiol Sci. 2001;16:1–5. doi: 10.1152/physiologyonline.2001.16.1.1. [DOI] [PubMed] [Google Scholar]
  • 6.Deaglio S, Robson SC. Ectonucleotidases as regulators of purinergic signaling in thrombosis, inflammation, and immunity. Adv Pharmacol. 2011;61:301–332. doi: 10.1016/B978-0-12-385526-8.00010-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Robson SC, Sevigny J, Zimmermann H. The E-NTPDase family of ectonucleotidases: structure function relationships and pathophysiological significance. Purinergic Signal. 2006;2:409–430. doi: 10.1007/s11302-006-9003-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Khakh BS, North RA. P2X receptors as cell-surface ATP sensors in health and disease. Nature. 2006;442:527–532. doi: 10.1038/nature04886. [DOI] [PubMed] [Google Scholar]
  • 9.Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat Rev Drug Discov. 2006;5:247–264. doi: 10.1038/nrd1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Erb L, Liao Z, Seye CI, Weisman GA. P2 receptors: intracellular signaling. Pflugers Arch. 2006;452:552–562. doi: 10.1007/s00424-006-0069-2. [DOI] [PubMed] [Google Scholar]
  • 11.Dussor G, Koerber HR, Oaklander AL, Rice FL, Molliver DC. Nucleotide signaling and cutaneous mechanisms of pain transduction. Brain Res Rev. 2009;60:24–35. doi: 10.1016/j.brainresrev.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ben Addi A, Cammarata D, Conley PB, Boeynaems JM, Robaye B. Role of the P2Y12 receptor in the modulation of murine dendritic cell function by ADP. J Immunol. 2010;185:5900–5906. doi: 10.4049/jimmunol.0901799. [DOI] [PubMed] [Google Scholar]
  • 13.Bassil AK, Bourdu S, Townson KA, Wheeldon A, Jarvie EM, Zebda N, Abuin A, Grau E, Livi GP, Punter L, et al. UDP-glucose modulates gastric function through P2Y14 receptor-dependent and -independent mechanisms. Am J Physiol Gastrointest Liver Physiol. 2009;296:G923–G930. doi: 10.1152/ajpgi.90363.2008. [DOI] [PubMed] [Google Scholar]
  • 14.Molliver DC, Rau KK, McIlwrath SL, Jankowski MP, Koerber HR. The ADP receptor P2Y1 is necessary for normal thermal sensitivity in cutaneous polymodal nociceptors. Mol Pain. 2011;7:13. doi: 10.1186/1744-8069-7-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Seye CI, Kong Q, Erb L, Garrad RC, Krugh B, Wang M, Turner JT, Sturek M, Gonzalez FA, Weisman GA. Functional P2Y2 nucleotide receptors mediate uridine 5′-triphosphate-induced intimal hyperplasia in collared rabbit carotid arteries. Circulation. 2002;106:2720–2726. doi: 10.1161/01.cir.0000038111.00518.35. [DOI] [PubMed] [Google Scholar]
  • 16.Blom D, Yamin TT, Champy MF, Selloum M, Bedu E, Carballo-Jane E, Gerckens L, Luell S, Meurer R, Chin J, et al. Altered lipoprotein metabolism in P2Y(13) knockout mice. Biochim Biophys Acta. 2010;1801:1349–1360. doi: 10.1016/j.bbalip.2010.08.013. [DOI] [PubMed] [Google Scholar]
  • 17.Wang N, Robaye B, Agrawal A, Skerry TM, Boeynaems JM, Gartland A. Reduced bone turnover in mice lacking the P2Y(13) receptor of ADP. Mol Endocrinol. 2012;26:142–152. doi: 10.1210/me.2011-1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Orriss IR, Utting JC, Brandao-Burch A, Colston K, Grubb BR, Burnstock G, Arnett TR. Extracellular nucleotides block bone mineralization in vitro: evidence for dual inhibitory mechanisms involving both P2Y2 receptors and pyrophosphate. Endocrinology. 2007;148:4208–4216. doi: 10.1210/en.2007-0066. [DOI] [PubMed] [Google Scholar]
  • 19.Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A, Nizet V, Insel PA, Junger WG. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314:1792–1795. doi: 10.1126/science.1132559. [DOI] [PubMed] [Google Scholar]
  • 20.Martin-Gil A, de Lara MJ, Crooke A, Santano C, Peral A, Pintor J. Silencing of P2Y(2) receptors reduces intraocular pressure in New Zealand rabbits. Br J Pharmacol. 2011;165:1163–1172. doi: 10.1111/j.1476-5381.2011.01586.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Muller T, Robaye B, Vieira RP, Ferrari D, Grimm M, Jakob T, Martin SF, Di Virgilio F, Boeynaems JM, Virchow JC, et al. The purinergic receptor P2Y2 receptor mediates chemotaxis of dendritic cells and eosinophils in allergic lung inflammation. Allergy. 2010;65:1545–1553. doi: 10.1111/j.1398-9995.2010.02426.x. [DOI] [PubMed] [Google Scholar]
  • 22.Cicko S, Lucattelli M, Muller T, Lommatzsch M, De Cunto G, Cardini S, Sundas W, Grimm M, Zeiser R, Durk T, et al. Purinergic receptor inhibition prevents the development of smoke-induced lung injury and emphysema. J Immunol. 2010;185:688–697. doi: 10.4049/jimmunol.0904042. [DOI] [PubMed] [Google Scholar]
  • 23.Matos JE, Robaye B, Boeynaems JM, Beauwens R, Leipziger J. K+ secretion activated by luminal P2Y2 and P2Y4 receptors in mouse colon. J Physiol. 2005;564:269–279. doi: 10.1113/jphysiol.2004.080002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bar I, Guns PJ, Metallo J, Cammarata D, Wilkin F, Boeynams JM, Bult H, Robaye B. Knockout mice reveal a role for P2Y6 receptor in macrophages, endothelial cells, and vascular smooth muscle cells. Mol Pharmacol. 2008;74:777–784. doi: 10.1124/mol.108.046904. [DOI] [PubMed] [Google Scholar]
  • 25.Orriss I, Syberg S, Wang N, Robaye B, Gartland A, Jorgensen N, Arnett T, Boeynaems JM. Bone phenotypes of P2 receptor knockout mice. Front Biosci (Schol Ed) 2011;3:1038–1046. doi: 10.2741/208. [DOI] [PubMed] [Google Scholar]
  • 26.Riegel AK, Faigle M, Zug S, Rosenberger P, Robaye B, Boeynaems JM, Idzko M, Eltzschig HK. Selective induction of endothelial P2Y6 nucleotide receptor promotes vascular inflammation. Blood. 2011;117:2548–2555. doi: 10.1182/blood-2010-10-313957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Laplante MA, Monassier L, Freund M, Bousquet P, Gachet C. The purinergic P2Y1 receptor supports leptin secretion in adipose tissue. Endocrinology. 2010;151:2060–2070. doi: 10.1210/en.2009-1134. [DOI] [PubMed] [Google Scholar]
  • 28.Hechler B, Freund M, Ravanat C, Magnenat S, Cazenave JP, Gachet C. Reduced atherosclerotic lesions in P2Y1/apolipoprotein E double-knockout mice: the contribution of non-hematopoietic-derived P2Y1 receptors. Circulation. 2008;118:754–763. doi: 10.1161/CIRCULATIONAHA.108.788927. [DOI] [PubMed] [Google Scholar]
  • 29.Rieg T, Bundey RA, Chen Y, Deschenes G, Junger W, Insel PA, Vallon V. Mice lacking P2Y2 receptors have salt-resistant hypertension and facilitated renal Na+ and water reabsorption. FASEB J. 2007;21:3717–3726. doi: 10.1096/fj.07-8807com. [DOI] [PubMed] [Google Scholar]
  • 30.Rumjahn SM, Yokdang N, Baldwin KA, Thai J, Buxton IL. Purinergic regulation of vascular endothelial growth factor signaling in angiogenesis. Br J Cancer. 2009;100:1465–1470. doi: 10.1038/sj.bjc.6604998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Boucher I, Rich C, Lee A, Marcincin M, Trinkaus-Randall V. The P2Y2 receptor mediates the epithelial injury response and cell migration. Am J Physiol Cell Physiol. 2010;299:C411–C421. doi: 10.1152/ajpcell.00100.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ward MM, Puthussery T, Fletcher EL. Localization and possible function of P2Y(4) receptors in the rodent retina. Neuroscience. 2008;155:1262–1274. doi: 10.1016/j.neuroscience.2008.06.035. [DOI] [PubMed] [Google Scholar]
  • 33.Guzman SJ, Schmidt H, Franke H, Krugel U, Eilers J, Illes P, Gerevich Z. P2Y1 receptors inhibit long-term depression in the prefrontal cortex. Neuropharmacology. 2010;59:406–415. doi: 10.1016/j.neuropharm.2010.05.013. [DOI] [PubMed] [Google Scholar]
  • 34.Kim B, Jeong HK, Kim JH, Lee SY, Jou I, Joe EH. Uridine 5′-diphosphate induces chemokine expression in microglia and astrocytes through activation of the P2Y6 receptor. J Immunol. 2011;186:3701–3709. doi: 10.4049/jimmunol.1000212. [DOI] [PubMed] [Google Scholar]
  • 35.Dorsam RT, Kunapuli SP. Central role of the P2Y12 receptor in platelet activation. J Clin Invest. 2004;113:340–345. doi: 10.1172/JCI20986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Vaughan KR, Stokes L, Prince LR, Marriott HM, Meis S, Kassack MU, Bingle CD, Sabroe I, Surprenant A, Whyte MK. Inhibition of neutrophil apoptosis by ATP is mediated by the P2Y11 receptor. J Immunol. 2007;179:8544–8553. doi: 10.4049/jimmunol.179.12.8544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Xiao Z, Yang M, Lv Q, Wang W, Deng M, Liu X, He Q, Chen X, Chen M, Fang L, et al. P2Y11 impairs cell proliferation by induction of cell cycle arrest and sensitizes endothelial cells to cisplatin-induced cell death. J Cell Biochem. 2011;112:2257–2265. doi: 10.1002/jcb.23144. [DOI] [PubMed] [Google Scholar]
  • 38.Arase T, Uchida H, Kajitani T, Ono M, Tamaki K, Oda H, Nishikawa S, Kagami M, Nagashima T, Masuda H, et al. The UDP-glucose receptor P2RY14 triggers innate mucosal immunity in the female reproductive tract by inducing IL-8. J Immunol. 2009;182:7074–7084. doi: 10.4049/jimmunol.0900001. [DOI] [PubMed] [Google Scholar]
  • 39.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9:1512–1519. doi: 10.1038/nn1805. [DOI] [PubMed] [Google Scholar]
  • 40.Wang L, Olivecrona G, Gotberg M, Olsson ML, Winzell MS, Erlinge D. ADP acting on P2Y13 receptors is a negative feedback pathway for ATP release from human red blood cells. Circ Res. 2005;96:189–196. doi: 10.1161/01.RES.0000153670.07559.E4. [DOI] [PubMed] [Google Scholar]
  • 41.Muller T, Bayer H, Myrtek D, Ferrari D, Sorichter S, Ziegenhagen MW, Zissel G, Virchow JC, Jr, Luttmann W, Norgauer J, et al. The P2Y14 receptor of airway epithelial cells: coupling to intracellular Ca2+ and IL-8 secretion. Am J Respir Cell Mol Biol. 2005;33:601–609. doi: 10.1165/rcmb.2005-0181OC. [DOI] [PubMed] [Google Scholar]
  • 42.Malin SA, Davis BM, Koerber HR, Reynolds IJ, Albers KM, Molliver DC. Thermal nociception and TRPV1 function are attenuated in mice lacking the nucleotide receptor P2Y2. Pain. 2008;138:484–496. doi: 10.1016/j.pain.2008.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Neves SR, Ram PT, Iyengar R. G protein pathways. Science. 2002;296:1636–1639. doi: 10.1126/science.1071550. [DOI] [PubMed] [Google Scholar]
  • 44.Waldo GL, Harden TK. Agonist binding and Gq-stimulating activities of the purified human P2Y1 receptor. Mol Pharmacol. 2004;65:426–436. doi: 10.1124/mol.65.2.426. [DOI] [PubMed] [Google Scholar]
  • 45.Bodor ET, Waldo GL, Hooks SB, Corbitt J, Boyer JL, Harden TK. Purification and functional reconstitution of the human P2Y12 receptor. Mol Pharmacol. 2003;64:1210–1216. doi: 10.1124/mol.64.5.1210. [DOI] [PubMed] [Google Scholar]
  • 46.Soulet C, Hechler B, Gratacap MP, Plantavid M, Offermanns S, Gachet C, Payrastre B. A differential role of the platelet ADP receptors P2Y1 and P2Y12 in Rac activation. J Thromb Haemost. 2005;3:2296–2306. doi: 10.1111/j.1538-7836.2005.01588.x. [DOI] [PubMed] [Google Scholar]
  • 47.Wallentin L. P2Y(12) inhibitors: differences in properties and mechanisms of action and potential consequences for clinical use. Eur Heart J. 2009;30:1964–1977. doi: 10.1093/eurheartj/ehp296. [DOI] [PubMed] [Google Scholar]
  • 48.Bagchi S, Liao Z, Gonzalez FA, Chorna NE, Seye CI, Weisman GA, Erb L. The P2Y2 nucleotide receptor interacts with alphav integrins to activate Go and induce cell migration. J Biol Chem. 2005;280:39050–39057. doi: 10.1074/jbc.M504819200. [DOI] [PubMed] [Google Scholar]
  • 49.Liao Z, Seye CI, Weisman GA, Erb L. The P2Y2 nucleotide receptor requires interaction with alpha v integrins to access and activate G12. J Cell Sci. 2007;120:1654–1662. doi: 10.1242/jcs.03441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Erb L, Liu J, Ockerhausen J, Kong Q, Garrad RC, Griffin K, Neal C, Krugh B, Santiago-Perez LI, Gonzalez FA, et al. An RGD sequence in the P2Y(2) receptor interacts with alpha(V)beta(3) integrins and is required for G(o)-mediated signal transduction. J Cell Biol. 2001;153:491–501. doi: 10.1083/jcb.153.3.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Baltensperger K, Porzig H. The P2U purinoceptor obligatorily engages the heterotrimeric G protein G16 to mobilize intracellular Ca2+ in human erythroleukemia cells. J Biol Chem. 1997;272:10151–10159. doi: 10.1074/jbc.272.15.10151. [DOI] [PubMed] [Google Scholar]
  • 52.Murthy KS, Makhlouf GM. Coexpression of ligand-gated P2X and G protein-coupled P2Y receptors in smooth muscle. Preferential activation of P2Y receptors coupled to phospholipase C (PLC)-beta1 via Galphaq/11 and to PLC-beta3 via Gbetagammai3. J Biol Chem. 1998;273:4695–4704. doi: 10.1074/jbc.273.8.4695. [DOI] [PubMed] [Google Scholar]
  • 53.Communi D, Motte S, Boeynaems JM, Pirotton S. Pharmacological characterization of the human P2Y4 receptor. Eur J Pharmacol. 1996;317:383–389. doi: 10.1016/s0014-2999(96)00740-6. [DOI] [PubMed] [Google Scholar]
  • 54.Nguyen T, Erb L, Weisman GA, Marchese A, Heng HH, Garrad RC, George SR, Turner JT, O'Dowd BF. Cloning, expression, and chromosomal localization of the human uridine nucleotide receptor gene. J Biol Chem. 1995;270:30845–30848. doi: 10.1074/jbc.270.52.30845. [DOI] [PubMed] [Google Scholar]
  • 55.Luykenaar KD, El-Rahman RA, Walsh MP, Welsh DG. Rho-kinase-mediated suppression of KDR current in cerebral arteries requires an intact actin cytoskeleton. Am J Physiol Heart Circ Physiol. 2009;296:H917–H926. doi: 10.1152/ajpheart.01206.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Filippov AK, Webb TE, Barnard EA, Brown DA. Dual coupling of heterologously-expressed rat P2Y6 nucleotide receptors to N-type Ca2+ and M-type K+ currents in rat sympathetic neurones. Br J Pharmacol. 1999;126:1009–1017. doi: 10.1038/sj.bjp.0702356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nishida M, Sato Y, Uemura A, Narita Y, Tozaki-Saitoh H, Nakaya M, Ide T, Suzuki K, Inoue K, Nagao T, et al. P2Y6 receptor-Galpha12/13 signalling in cardiomyocytes triggers pressure overload-induced cardiac fibrosis. EMBO J. 2008;27:3104–3115. doi: 10.1038/emboj.2008.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kaufmann A, Musset B, Limberg SH, Renigunta V, Sus R, Dalpke AH, Heeg KM, Robaye B, Hanley PJ. “Host tissue damage” signal ATP promotes non-directional migration and negatively regulates toll-like receptor signaling in human monocytes. J Biol Chem. 2005;280:32459–32467. doi: 10.1074/jbc.M505301200. [DOI] [PubMed] [Google Scholar]
  • 59.Nguyen TD, Meichle S, Kim US, Wong T, Moody MW. P2Y(11), a purinergic receptor acting via cAMP, mediates secretion by pancreatic duct epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2001;280:G795–G804. doi: 10.1152/ajpgi.2001.280.5.G795. [DOI] [PubMed] [Google Scholar]
  • 60.White PJ, Webb TE, Boarder MR. Characterization of a Ca2+ response to both UTP and ATP at human P2Y11 receptors: evidence for agonist-specific signaling. Mol Pharmacol. 2003;63:1356–1363. doi: 10.1124/mol.63.6.1356. [DOI] [PubMed] [Google Scholar]
  • 61.Ecke D, Hanck T, Tulapurkar ME, Schafer R, Kassack M, Stricker R, Reiser G. Hetero-oligomerization of the P2Y11 receptor with the P2Y1 receptor controls the internalization and ligand selectivity of the P2Y11 receptor. Biochem J. 2008;409:107–116. doi: 10.1042/BJ20070671. [DOI] [PubMed] [Google Scholar]
  • 62.Soulet C, Sauzeau V, Plantavid M, Herbert JM, Pacaud P, Payrastre B, Savi P. Gi-dependent and -independent mechanisms downstream of the P2Y12 ADP-receptor. J Thromb Haemost. 2004;2:135–146. doi: 10.1111/j.1538-7836.2004.00556.x. [DOI] [PubMed] [Google Scholar]
  • 63.Van Kolen K, Slegers H. Atypical PKCzeta is involved in RhoA-dependent mitogenic signaling by the P2Y(12) receptor in C6 cells. FEBS J. 2006;273:1843–1854. doi: 10.1111/j.1742-4658.2006.05205.x. [DOI] [PubMed] [Google Scholar]
  • 64.Marteau F, Le Poul E, Communi D, Labouret C, Savi P, Boeynaems JM, Gonzalez NS. Pharmacological characterization of the human P2Y13 receptor. Mol Pharmacol. 2003;64:104–112. doi: 10.1124/mol.64.1.104. [DOI] [PubMed] [Google Scholar]
  • 65.Malaval C, Laffargue M, Barbaras R, Rolland C, Peres C, Champagne E, Perret B, Terce F, Collet X, Martinez LO. RhoA/ROCK I signalling downstream of the P2Y13 ADP-receptor controls HDL endocytosis in human hepatocytes. Cell Signal. 2009;21:120–127. doi: 10.1016/j.cellsig.2008.09.016. [DOI] [PubMed] [Google Scholar]
  • 66.Harden TK, Sesma JI, Fricks IP, Lazarowski ER. Signalling and pharmacological properties of the P2Y receptor. Acta Physiol (Oxf) 2010;199:149–160. doi: 10.1111/j.1748-1716.2010.02116.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yu N, Erb L, Shivaji R, Weisman GA, Seye CI. Binding of the P2Y2 nucleotide receptor to filamin A regulates migration of vascular smooth muscle cells. Circ Res. 2008;102:581–588. doi: 10.1161/CIRCRESAHA.107.162271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Suzuki T, Namba K, Tsuga H, Nakata H. Regulation of pharmacology by hetero-oligomerization between A1 adenosine receptor and P2Y2 receptor. Biochem Biophys Res Commun. 2006;351:559–565. doi: 10.1016/j.bbrc.2006.10.075. [DOI] [PubMed] [Google Scholar]
  • 69.Yoshioka K, Saitoh O, Nakata H. Heteromeric association creates a P2Y-like adenosine receptor. Proc Natl Acad Sci USA. 2001;98:7617–7622. doi: 10.1073/pnas.121587098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shrestha SS, Parmar M, Kennedy C, Bushell TJ. Two-pore potassium ion channels are inhibited by both G(q/11)- and G(i)-coupled P2Y receptors. Mol Cell Neurosci. 2010;43:363–369. doi: 10.1016/j.mcn.2010.01.003. [DOI] [PubMed] [Google Scholar]
  • 71.Savi P, Zachayus JL, Delesque-Touchard N, Labouret C, Herve C, Uzabiaga MF, Pereillo JM, Culouscou JM, Bono F, Ferrara P, et al. The active metabolite of Clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts. Proc Natl Acad Sci USA. 2006;103:11069–11074. doi: 10.1073/pnas.0510446103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.D'Ambrosi N, Iafrate M, Saba E, Rosa P, Volonte C. Comparative analysis of P2Y4 and P2Y6 receptor architecture in native and transfected neuronal systems. Biochim Biophys Acta. 2007;1768:1592–1599. doi: 10.1016/j.bbamem.2007.03.020. [DOI] [PubMed] [Google Scholar]
  • 73.Ng SW, Bakowski D, Nelson C, Mehta R, Almeyda R, Bates G, Parekh AB. Cysteinyl leukotriene type I receptor desensitization sustains Ca2+-dependent gene expression. Nature. 2012;482:111–115. doi: 10.1038/nature10731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Shenoy SK, Lefkowitz RJ. beta-Arrestin-mediated receptor trafficking and signal transduction. Trends Pharmacol Sci. 2011;32:521–533. doi: 10.1016/j.tips.2011.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hoffmann C, Ziegler N, Reiner S, Krasel C, Lohse MJ. Agonist-selective, receptor-specific interaction of human P2Y receptors with beta-arrestin-1 and -2. J Biol Chem. 2008;283:30933–30941. doi: 10.1074/jbc.M801472200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Velazquez B, Garrad RC, Weisman GA, Gonzalez FA. Differential agonist-induced desensitization of P2Y2 nucleotide receptors by ATP and UTP. Mol Cell Biochem. 2000;206:75–89. doi: 10.1023/a:1007091127392. [DOI] [PubMed] [Google Scholar]
  • 77.Morris GE, Nelson CP, Brighton PJ, Standen NB, Challiss RA, Willets JM. Arrestins differentially regulate ETA and P2Y2 receptor-mediated cell signaling and migration in arterial smooth muscle. Am J Physiol Cell Physiol. 2012;302:C723–C734. doi: 10.1152/ajpcell.00202.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Morris GE, Nelson CP, Everitt D, Brighton PJ, Standen NB, Challiss RA, Willets JM. G protein-coupled receptor kinase 2 and arrestin2 regulate arterial smooth muscle P2Y-purinoceptor signalling. Cardiovasc Res. 2011;89:193–203. doi: 10.1093/cvr/cvq249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Garrad RC, Otero MA, Erb L, Theiss PM, Clarke LL, Gonzalez FA, Turner JT, Weisman GA. Structural basis of agonist-induced desensitization and sequestration of the P2Y2 nucleotide receptor. Consequences of truncation of the C terminus. J Biol Chem. 1998;273:29437–29444. doi: 10.1074/jbc.273.45.29437. [DOI] [PubMed] [Google Scholar]
  • 80.Flores RV, Hernandez-Perez MG, Aquino E, Garrad RC, Weisman GA, Gonzalez FA. Agonist-induced phosphorylation and desensitization of the P2Y2 nucleotide receptor. Mol Cell Biochem. 2005;280:35–45. doi: 10.1007/s11010-005-8050-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hardy AR, Conley PB, Luo J, Benovic JL, Poole AW, Mundell SJ. P2Y1 and P2Y12 receptors for ADP desensitize by distinct kinase-dependent mechanisms. Blood. 2005;105:3552–3560. doi: 10.1182/blood-2004-07-2893. [DOI] [PubMed] [Google Scholar]
  • 82.Nisar S, Daly ME, Federici AB, Artoni A, Mumford AD, Watson SP, Mundell SJ. An intact PDZ motif is essential for correct P2Y12 purinoceptor traffic in human platelets. Blood. 2011;118:5641–5651. doi: 10.1182/blood-2011-02-336826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wolff SC, Qi AD, Harden TK, Nicholas RA. Polarized expression of human P2Y receptors in epithelial cells from kidney, lung, and colon. Am J Physiol Cell Physiol. 2005;288:C624–C632. doi: 10.1152/ajpcell.00338.2004. [DOI] [PubMed] [Google Scholar]
  • 84.Wolff SC, Qi AD, Harden TK, Nicholas RA. Charged residues in the C-terminus of the P2Y1 receptor constitute a basolateral-sorting signal. J Cell Sci. 2010;123:2512–2520. doi: 10.1242/jcs.060723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Qi AD, Wolff SC, Nicholas RA. The apical targeting signal of the P2Y2 receptor is located in its first extra-cellular loop. J Biol Chem. 2005;280:29169–29175. doi: 10.1074/jbc.M501301200. [DOI] [PubMed] [Google Scholar]
  • 86.Ebisuya M, Kondoh K, Nishida E. The duration, magnitude and compartmentalization of ERK MAP kinase activity: mechanisms for providing signaling specificity. J Cell Sci. 2005;118:2997–3002. doi: 10.1242/jcs.02505. [DOI] [PubMed] [Google Scholar]
  • 87.Ichikawa J, Furuya K, Miyata S, Nakashima T, Kiyohara T. EGF enhances Ca(2+) mobilization and capacitative Ca(2+) entry in mouse mammary epithelial cells. Cell Biochem Funct. 2000;18:215–225. doi: 10.1002/1099-0844(200009)18:3<215::AID-CBF875>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 88.Gutierrez AM, Lou X, Erik A, Persson G, Ring A. Growth hormones reverse desensitization of P2Y(2) receptors in rat mesangial cells. Biochem Biophys Res Commun. 2000;270:594–599. doi: 10.1006/bbrc.2000.2461. [DOI] [PubMed] [Google Scholar]
  • 89.Weick M, Wiedemann P, Reichenbach A, Bringmann A. Resensitization of P2Y receptors by growth factor-mediated activation of the phosphatidylinositol-3 kinase in retinal glial cells. Invest Ophthalmol Vis Sci. 2005;46:1525–1532. doi: 10.1167/iovs.04-0417. [DOI] [PubMed] [Google Scholar]
  • 90.Marchese A, Paing MM, Temple BR, Trejo J. G protein-coupled receptor sorting to endosomes and lysosomes. Annu Rev Pharmacol Toxicol. 2008;48:601–629. doi: 10.1146/annurev.pharmtox.48.113006.094646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Gerwins P, Fredholm BB. ATP and its metabolite adenosine act synergistically to mobilize intracellular calcium via the formation of inositol 1,4,5-trisphosphate in a smooth muscle cell line. J Biol Chem. 1992;267:16081–16087. [PubMed] [Google Scholar]
  • 92.Megson AC, Dickenson JM, Townsend-Nicholson A, Hill SJ. Synergy between the inositol phosphate responses to transfected human adenosine A1-receptors and constitutive P2-purinoceptors in CHO-K1 cells. Br J Pharmacol. 1995;115:1415–1424. doi: 10.1111/j.1476-5381.1995.tb16632.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Selbie LA, King NV, Dickenson JM, Hill SJ. Role of G-protein beta gamma subunits in the augmentation of P2Y2 (P2U)receptor-stimulated responses by neuropeptide Y Y1 Gi/o-coupled receptors. Biochem J. 1997;328(Pt 1):153–158. doi: 10.1042/bj3280153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Werry TD, Christie MI, Dainty IA, Wilkinson GF, Willars GB. Ca(2+) signalling by recombinant human CXCR2 chemokine receptors is potentiated by P2Y nucleotide receptors in HEK cells. Br J Pharmacol. 2002;135:1199–1208. doi: 10.1038/sj.bjp.0704566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Felder CC, Williams HL, Axelrod J. A transduction pathway associated with receptors coupled to the inhibitory guanine nucleotide bindinG protein Gi that amplifies ATP-mediated arachidonic acid release. Proc Natl Acad Sci USA. 1991;88:6477–6480. doi: 10.1073/pnas.88.15.6477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wang H, Wang DH, Galligan JJ. P2Y2 receptors mediate ATP-induced resensitization of TRPV1 expressed by kidney projecting sensory neurons. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1634–1641. doi: 10.1152/ajpregu.00235.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Tominaga M, Wada M, Masu M. Potentiation of capsaicin receptor activity by metabotropic ATP receptors as a possible mechanism for ATP-evoked pain and hyperalgesia. Proc Natl Acad Sci USA. 2001;98:6951–6956. doi: 10.1073/pnas.111025298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Vial C, Tobin AB, Evans RJ. G-protein-coupled receptor regulation of P2X1 receptors does not involve direct channel phosphorylation. Biochem J. 2004;382:101–110. doi: 10.1042/BJ20031910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vial C, Rolf MG, Mahaut-Smith MP, Evans RJ. A study of P2X1 receptor function in murine megakaryocytes and human platelets reveals synergy with P2Y receptors. Br J Pharmacol. 2002;135:363–372. doi: 10.1038/sj.bjp.0704486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tolhurst G, Vial C, Leon C, Gachet C, Evans RJ, Mahaut-Smith MP. Interplay between P2Y(1), P2Y(12), and P2X(1) receptors in the activation of megakaryocyte cation influx currents by ADP: evidence that the primary megakaryocyte represents a fully functional model of platelet P2 receptor signaling. Blood. 2005;106:1644–1651. doi: 10.1182/blood-2005-02-0725. [DOI] [PubMed] [Google Scholar]
  • 101.Chen X, Molliver DC, Gebhart GF. The P2Y2 receptor sensitizes mouse bladder sensory neurons and facilitates purinergic currents. J Neurosci. 2010;30:2365–2372. doi: 10.1523/JNEUROSCI.5462-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dubyak GR. Knock-out mice reveal tissue-specific roles of P2Y receptor subtypes in different epithelia. Mol Pharmacol. 2003;63:773–776. doi: 10.1124/mol.63.4.773. [DOI] [PubMed] [Google Scholar]
  • 103.Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL, Kennedy C, Knight GE, Fumagalli M, Gachet C, Jacobson KA, et al. International Union of Pharmacology LVIII: update on the P2Y G protein-coupled nucleotide receptors: from molecular mechanisms and pathophysiology to therapy. Pharmacol Rev. 2006;58:281–341. doi: 10.1124/pr.58.3.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Cressman VL, Lazarowski E, Homolya L, Boucher RC, Koller BH, Grubb BR. Effect of loss of P2Y(2) receptor gene expression on nucleotide regulation of murine epithelial Cl(−) transport. J Biol Chem. 1999;274:26461–26468. doi: 10.1074/jbc.274.37.26461. [DOI] [PubMed] [Google Scholar]
  • 105.Robaye B, Ghanem E, Wilkin F, Fokan D, Van Driessche W, Schurmans S, Boeynaems JM, Beauwens R. Loss of nucleotide regulation of epithelial chloride transport in the jejunum of P2Y4-null mice. Mol Pharmacol. 2003;63:777–783. doi: 10.1124/mol.63.4.777. [DOI] [PubMed] [Google Scholar]
  • 106.Hall RA, Ostedgaard LS, Premont RT, Blitzer JT, Rahman N, Welsh MJ, Lefkowitz RJ. A C-terminal motif found in the beta2-adrenergic receptor, P2Y1 receptor and cystic fibrosis transmembrane conductance regulator determines binding to the Na+/H+ exchanger regulatory factor family of PDZ proteins. Proc Natl Acad Sci USA. 1998;95:8496–8501. doi: 10.1073/pnas.95.15.8496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lee SY, Wolff SC, Nicholas RA, O'Grady SM. P2Y receptors modulate ion channel function through interactions involving the C-terminal domain. Mol Pharmacol. 2003;63:878–885. doi: 10.1124/mol.63.4.878. [DOI] [PubMed] [Google Scholar]
  • 108.Bahra P, Mesher J, Li S, Poll CT, Danahay H. P2Y2-receptor-mediated activation of a contralateral, lanthanide-sensitive calcium entry pathway in the human airway epithelium. Br J Pharmacol. 2004;143:91–98. doi: 10.1038/sj.bjp.0705913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Viana F, de Smedt H, Droogmans G, Nilius B. Calcium signalling through nucleotide receptor P2Y2 in cultured human vascular endothelium. Cell Calcium. 1998;24:117–127. doi: 10.1016/s0143-4160(98)90079-3. [DOI] [PubMed] [Google Scholar]
  • 110.Dong X, Smoll EJ, Ko KH, Lee J, Chow JY, Kim HD, Insel PA, Dong H. P2Y receptors mediate Ca2+ signaling in duodenocytes and contribute to duodenal mucosal bicarbonate secretion. Am J Physiol Gastrointest Liver Physiol. 2009;296:G424–G432. doi: 10.1152/ajpgi.90314.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kellerman D, Evans R, Mathews D, Shaffer C. Inhaled P2Y2 receptor agonists as a treatment for patients with Cystic Fibrosis lung disease. Adv Drug Deliv Rev. 2002;54:1463–1474. doi: 10.1016/s0169-409x(02)00154-0. [DOI] [PubMed] [Google Scholar]
  • 112.Paradiso AM, Ribeiro CM, Boucher RC. Polarized signaling via purinoceptors in normal and cystic fibrosis airway epithelia. J Gen Physiol. 2001;117:53–67. doi: 10.1085/jgp.117.1.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Parr CE, Sullivan DM, Paradiso AM, Lazarowski ER, Burch LH, Olsen JC, Erb L, Weisman GA, Boucher RC, Turner JT. Cloning and expression of a human P2U nucleotide receptor, a target for cystic fibrosis pharmacotherapy. Proc Natl Acad Sci USA. 1994;91:3275–3279. doi: 10.1073/pnas.91.8.3275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Yerxa BR, Sabater JR, Davis CW, Stutts MJ, Lang-Furr M, Picher M, Jones AC, Cowlen M, Dougherty R, Boyer J, et al. Pharmacology of INS37217 [P(1)-(uridine 5′)-P(4)- (2′-deoxycytidine 5′)tetraphosphate, tetrasodium salt], a next-generation P2Y(2) receptor agonist for the treatment of cystic fibrosis. J Pharmacol Exp Ther. 2002;302:871–880. doi: 10.1124/jpet.102.035485. [DOI] [PubMed] [Google Scholar]
  • 115.Meyer CH, Hotta K, Peterson WM, Toth CA, Jaffe GJ. Effect of INS37217, a P2Y(2) receptor agonist, on experimental retinal detachment and electroretinogram in adult rabbits. Invest Ophthalmol Vis Sci. 2002;43:3567–3574. [PubMed] [Google Scholar]
  • 116.Kim CH, Kim HY, Lee HS, Chang SO, Oh SH, Lee JH. P2Y4-mediated regulation of Na+ absorption in the Reissner's membrane of the cochlea. J Neurosci. 2010;30:3762–3769. doi: 10.1523/JNEUROSCI.3300-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Marcus DC, Liu J, Lee JH, Scherer EQ, Scofield MA, Wangemann P. Apical membrane P2Y4 purinergic receptor controls K+ secretion by strial marginal cell epithelium. Cell Commun Signal. 2005;3:13. doi: 10.1186/1478-811X-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Filippov AK, Simon J, Barnard EA, Brown DA. Coupling of the nucleotide P2Y4 receptor to neuronal ion channels. Br J Pharmacol. 2003;138:400–406. doi: 10.1038/sj.bjp.0705043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Malin SA, Molliver DC. Gi- and Gq-coupled ADP (P2Y) receptors act in opposition to modulate nociceptive signaling and inflammatory pain behavior. Mol Pain. 2010;6:21. doi: 10.1186/1744-8069-6-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Marker CL, Lujan R, Loh HH, Wickman K. Spinal G-protein-gated potassium channels contribute in a dose-dependent manner to the analgesic effect of mu-and delta- but not kappa-opioids. J Neurosci. 2005;25:3551–3559. doi: 10.1523/JNEUROSCI.4899-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Filippov AK, Fernandez-Fernandez JM, Marsh SJ, Simon J, Barnard EA, Brown DA. Activation and inhibition of neuronal G protein-gated inwardly rectifying K(+) channels by P2Y nucleotide receptors. Mol Pharmacol. 2004;66:468–477. doi: 10.1124/mol.66.3.. [DOI] [PubMed] [Google Scholar]
  • 122.Wu J, Ding WG, Matsuura H, Horie M. Regulatory mechanisms underlying the modulation of GIRK1/GIRK4 heteromeric channels by P2Y receptors. Pflugers Arch. 2012;463:625–633. doi: 10.1007/s00424-012-1082-2. [DOI] [PubMed] [Google Scholar]
  • 123.Mathie A. Neuronal two-pore-domain potassium channels and their regulation by G protein-coupled receptors. J Physiol. 2007;578:377–385. doi: 10.1113/jphysiol.2006.121582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Liu J, Liao Z, Camden J, Griffin KD, Garrad RC, Santiago-Perez LI, Gonzalez FA, Seye CI, Weisman GA, Erb L. Src homology 3 binding sites in the P2Y2 nucleotide receptor interact with Src and regulate activities of Src, proline-rich tyrosine kinase 2, and growth factor receptors. J Biol Chem. 2004;279:8212–8218. doi: 10.1074/jbc.M312230200. [DOI] [PubMed] [Google Scholar]
  • 125.Seye CI, Yu N, Gonzalez FA, Erb L, Weisman GA. The P2Y2 nucleotide receptor mediates vascular cell adhesion molecule-1 expression through interaction with VEGF receptor-2 (KDR/Flk-1) J Biol Chem. 2004;279:35679–35686. doi: 10.1074/jbc.M401799200. [DOI] [PubMed] [Google Scholar]
  • 126.Ratchford AM, Baker OJ, Camden JM, Rikka S, Petris MJ, Seye CI, Erb L, Weisman GA. P2Y2 nucleotide receptors mediate metalloprotease-dependent phosphorylation of epidermal growth factor receptor and ErbB3 in human salivary gland cells. J Biol Chem. 2010;285:7545–7555. doi: 10.1074/jbc.M109.078170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Yin J, Xu K, Zhang J, Kumar A, Yu FS. Wound-induced ATP release and EGF receptor activation in epithelial cells. J Cell Sci. 2007;120:815–825. doi: 10.1242/jcs.03389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Van Kolen K, Gilany K, Moens L, Esmans EL, Slegers H. P2Y12 receptor signalling towards PKB proceeds through IGF-I receptor cross-talk and requires activation of Src, Pyk2 and Rap1. Cell Signal. 2006;18:1169–1181. doi: 10.1016/j.cellsig.2005.09.005. [DOI] [PubMed] [Google Scholar]

RESOURCES