+ All Categories
Home > Documents > Purinergic signaling and kinase activation for survival in pulmonary oxidative stress and disease

Purinergic signaling and kinase activation for survival in pulmonary oxidative stress and disease

Date post: 04-Sep-2016
Category:
Upload: shama-ahmad
View: 217 times
Download: 3 times
Share this document with a friend
12
Serial Review: Redox Signaling in Immune Function and Cellular Responses in Lung Injury and Diseases Serial Review Editors: Victor DarleyUsmar, Lin Mantell Purinergic signaling and kinase activation for survival in pulmonary oxidative stress and disease Shama Ahmad , Aftab Ahmad, Carl W. White Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson Street, Denver, CO 80206, USA Received 11 November 2005; revised 27 February 2006; accepted 2 March 2006 Available online 31 March 2006 Abstract Stimulus-induced release of endogenous ATP into the extracellular milieu has been shown to occur in a variety of cells, tissues, and organs. Extracellular ATP can propagate signals via P2 receptors that are essential for growth and survival of cells. Abundance of P2 receptors, their multiple isoforms, and their ubiquitous distribution indicate that they transmit vital signals. Pulmonary epithelium and endothelium are rich in both P2X and P2Y receptors. ATP release from lung tissue and cells occurs upon stimulation both in vivo and in vitro. Extracellular ATP can activate signaling cascades composed of protein kinases including extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3-kinase (PI3K). Here we summarize progress related to release of endogenous ATP and nucleotide signaling in pulmonary tissues upon exposure to oxidant stress. Hypoxic, hyperoxic, and ozone exposures cause a rapid increase of extracellular ATP in primary pulmonary endothelial and epithelial cells. Extracellular ATP is critical for survival of these cells in high oxygen and ozone concentrations. The released ATP, upon binding to its specific receptors, triggers ERK and PI3K signaling and renders cells resistant to these stresses. Impairment of ATP release and transmission of such signals could limit cellular survival under oxidative stress. This may further contribute to disease pathogenesis or exacerbation. © 2006 Elsevier Inc. All rights reserved. Keywords: ATP; Nucleotide receptors; Oxidant; Pulmonary; Cystic fibrosis; Protein kinase; Free radical Contents Introduction ................................................................. 30 Purinoreceptor expression in pulmonary cells ............................................... 31 Endogenous ATP release in lung and its effects .............................................. 32 ATP-mediated protection against oxidative stress .............................................. 34 Signaling events associated with ATP-mediated protection ......................................... 34 Acknowledgments ............................................................. 36 References ................................................................. 36 Free Radical Biology & Medicine 41 (2006) 29 40 www.elsevier.com/locate/freeradbiomed Abbreviations: BPD, bronchopulmonary dysplasia; CFTR, cystic fibrosis transmembrane conductance regulator; FKBP12, FK506-binding protein; ERK, extracellular signal-regulated kinase; HLMVEC, human lung microvascular endothelial cells; HKII, hexokinase II; IP 3 , inositol trisphosphate; LDH, lactate dehydrogenase; MAPK, mitogen-activated protein kinases; MEK, mitogen-activated protein kinase kinase or ERK kinase; MDR, multidrug resistance; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol 3-kinase; PLA 2 , phospholipase A 2 ; PLD, phospholipase D; PLC, phospholipase C; PKC, protein kinase C; P70S6K, ribosomal p70 S6 kinase; PGE 2 , prostaglandin E 2 ; RTPCR, reverse-transcriptase polymerase chain reaction; ROS, reactive oxygen species; RVD, regulatory volume decrease; VDAC, voltage-dependent anion channel. This article is part of a series of reviews on Redox Signaling in Immune Function and Cellular Responses in Lung Injury and Diseases.The full list of papers may be found on the home page of the journal. Corresponding author. Fax: +1 303 398 1851. E-mail address: [email protected] (S. Ahmad). 0891-5849/$ - see front matter © 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.freeradbiomed.2006.03.004
Transcript

Free Radical Biology & Medicine 41 (2006) 29–40www.elsevier.com/locate/freeradbiomed

Serial Review: Redox Signaling in Immune Function and Cellular Responses in Lung Injury and DiseasesSerial Review Editors: Victor Darley–Usmar, Lin Mantell

Purinergic signaling and kinase activation for survival in pulmonaryoxidative stress and disease☆

Shama Ahmad ⁎, Aftab Ahmad, Carl W. White

Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson Street, Denver, CO 80206, USA

Received 11 November 2005; revised 27 February 2006; accepted 2 March 2006Available online 31 March 2006

Abstract

Stimulus-induced release of endogenous ATP into the extracellular milieu has been shown to occur in a variety of cells, tissues, and organs.Extracellular ATP can propagate signals via P2 receptors that are essential for growth and survival of cells. Abundance of P2 receptors, theirmultiple isoforms, and their ubiquitous distribution indicate that they transmit vital signals. Pulmonary epithelium and endothelium are rich in bothP2X and P2Y receptors. ATP release from lung tissue and cells occurs upon stimulation both in vivo and in vitro. Extracellular ATP can activatesignaling cascades composed of protein kinases including extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3-kinase (PI3K).Here we summarize progress related to release of endogenous ATP and nucleotide signaling in pulmonary tissues upon exposure to oxidant stress.Hypoxic, hyperoxic, and ozone exposures cause a rapid increase of extracellular ATP in primary pulmonary endothelial and epithelial cells.Extracellular ATP is critical for survival of these cells in high oxygen and ozone concentrations. The released ATP, upon binding to its specificreceptors, triggers ERK and PI3K signaling and renders cells resistant to these stresses. Impairment of ATP release and transmission of suchsignals could limit cellular survival under oxidative stress. This may further contribute to disease pathogenesis or exacerbation.© 2006 Elsevier Inc. All rights reserved.

Keywords: ATP; Nucleotide receptors; Oxidant; Pulmonary; Cystic fibrosis; Protein kinase; Free radical

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30Purinoreceptor expression in pulmonary cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31Endogenous ATP release in lung and its effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32ATP-mediated protection against oxidative stress. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34Signaling events associated with ATP-mediated protection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36

Abbreviations: BPD, bronchopulmonary dysplasia; CFTR, cystic fibrosis transmembrane conductance regulator; FKBP12, FK506-binding protein; ERK,extracellular signal-regulated kinase; HLMVEC, human lung microvascular endothelial cells; HKII, hexokinase II; IP3, inositol trisphosphate; LDH, lactatedehydrogenase; MAPK, mitogen-activated protein kinases; MEK, mitogen-activated protein kinase kinase or ERK kinase; MDR, multidrug resistance; mTOR,mammalian target of rapamycin; PI3K, phosphatidylinositol 3-kinase; PLA2, phospholipase A2; PLD, phospholipase D; PLC, phospholipase C; PKC, protein kinaseC; P70S6K, ribosomal p70 S6 kinase; PGE2, prostaglandin E2; RTPCR, reverse-transcriptase polymerase chain reaction; ROS, reactive oxygen species; RVD,regulatory volume decrease; VDAC, voltage-dependent anion channel.☆ This article is part of a series of reviews on “Redox Signaling in Immune Function and Cellular Responses in Lung Injury and Diseases.” The full list of papers maybe found on the home page of the journal.⁎ Corresponding author. Fax: +1 303 398 1851.E-mail address: [email protected] (S. Ahmad).

0891-5849/$ - see front matter © 2006 Elsevier Inc. All rights reserved.doi:10.1016/j.freeradbiomed.2006.03.004

30 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

Introduction

ATP is the principal intracellular energy source. Recentattention has been focused on ATP's functions outside the cell.Cytosolic ATP and that packed into secretory granules andvesicles can be released from cells either under basal conditionsor upon stimulation by cellular stress via regulated nonlyticmechanisms [1,2]. Released ATP and its metabolites ADP andadenosine can mediate a diverse spectrum of effects in manycell types upon binding to their distinct and specific receptors.P1 receptors are activated by adenosine, whereas ATP and itsanalogs activate P2 receptors. P2 receptors are of two types: theionotropic P2X and the metabotropic P2Y. Cells expressmultiple isoforms of P2X and P2Y receptors (Fig. 1) [2,3].Once released, ATP interacts with these purinergic receptorsand activates rapid signaling events that are both autocrine andparacrine [4]. Thus, ATP is an important signaling molecule thatparticipates in intercellular communication and also regulates abroad range of physiological responses like (i) increases inintracellular calcium; (ii) activation of phospholipases; (iii)modulation of cell volume; (iv) inhibition of platelet aggrega-tion; (v) alteration of vascular tone; (vi) neurotransmission; (vii)elevation of cardiac/skeletal muscle contractility; (viii) immunecell activation; (ix) increased ciliary beat frequency and mucussecretion; (x) pulmonary surfactant release from alveolar type IIepithelial cells; (xi) bronchoconstriction in individuals withhyperreactive airways; and (xii) enhanced cell growth andproliferation [5]. Extracellular ATP and adenosine may alsocause cell death/apoptosis depending on their extracellularconcentration and cell type so exposed [6].

ATP release and ATP-mediated signaling during oxidativestress have been less thoroughly investigated in the lung than inthe cardiovascular system. Several studies indicate thatpurinergic receptors are involved in the cellular response tooxidative stress [7–13]. However, the role of ATP release in

Fig. 1. Mammalian purine receptors. There are two main families of purine receptorsand UDP. P1 receptors comprise A1, A2A, A2B, and A3 receptors, and P2 receptors a(GPCR). Their structure consists of typical features of GPCRs including 7 transmembamino acid residues in their transmembrane region determine the ligand binding and sA3 receptors. P2Y receptors are further subclassified into two phylogenetically dist(P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11), and the other containing receptors that couphighly diverse in both their amino acid sequences (<18% sequence homology) and punrelated in amino acid sequence to other ion channels and other proteins, but sharsubtypes P2X1–P2X7 which often oligomerize to form heteromers and homomers. T

oxidative stress has not been altogether clear. Severe oxidativestress can diminish intracellular ATP [14,15]. Delayed catab-olism of extracellular nucleotides due to diminished ecto-nucleotidase activity in the presence of reactive oxygen species(ROS) also has been suggested [15]. Further, extracellular ATPitself could lead to oxidative stress in some cell types [16,17].For example, extracellular ATP can induce H2O2 production byincreasing Ca2+-mediated Duox, an NADPH oxidase, in airwayepithelium [18]. Stimulation of P2X receptors also can inducesuperoxide production and contraction of mouse aorta [19].Thus, ATP not only responds to redox changes but also canitself trigger redox changes.

Oxidative stress like that in hyperoxia also signals throughvarious pathways. Previously we and others have shown thatmicrovascular endothelial cells are primary targets of hyperoxiclung injury and in other forms of acute respiratory failure[20,21]. Hypoxic preexposure of human lung microvascularendothelial cells (HLMVEC) attenuates cell death caused bysubsequent hyperoxic exposure [21]. This is partially attributedto upregulation of PI 3-kinase (PI3K), a component of anestablished cell survival pathway, during the hyperoxicexposure. Hypoxic (1 or 3% O2) exposure also induces ATPrelease in HLMVEC and fetal lung fibroblasts [22,23]. There,hypoxia and extracellular ATP acted synergistically to upregu-late common proliferation-inducing pathways in lung adventi-tial fibroblasts. Extracellular ATP and purinergic receptoractivation play a critical role in determining the intracellulareffects of key growth-regulating factors [4,24,25]. In addition toATP release, hypoxic conditions also favor proliferation inendothelial cells [22,26]. In primary HLMVEC, we have alsoshown previously that ATP is released in hyperoxia and that thepresence of extracellular ATP is essential for survival of thesecells [27]. Enhanced glucose uptake and upregulation ofhexokinase II (HKII) are other potential contributing survivalfactors in hyperoxia [28,29]. A possible link between these

: adenosine or P1 receptors and P2 receptors, which recognize ATP, ADP, UTP,re of two types the P2X and P2Y. P1 and P2Y are G protein-coupled receptorsrane regions connected by 3 extracellular and 3 intracellular loops. The conservedpecificity. P1 receptors share <50% homology among cloned A1, A2A, A2B, andinct groups, one encompassing the receptors that mainly couple to Gq proteinsle to Gi proteins (P2Y12, P2Y13, and P2Y14). The mammalian P2Y subtypes areharmacological profiles. P2X receptors are ligand-gated ion channels. They aree striking topological similarity to epithelial sodium channels. There are sevenhe P2X receptor proteins share ∼30–50% sequence identity.

Table 2P2Y receptor distribution in lungs

Receptorsubtype

Tissue/species Method of detection References

P2Y1 16HBE14o-/human Confocal microscopy [144]Lung parenchyma/rabbit RTPCR [145]Pulmonary artery/rabbitPulmonary artery ECs/rabbitCalu3 cells/human Northern blot [146]

RTPCR [147]Antagonist reactivity

Pulmonary artery ECs/bovine Agonist reactivity [148][149]

Airway epithelial cells/mice Agonist reactivity [128]Airway smooth musclecells/human

RTPCR [150]

P2Y2 Microvascular ECs/human Agonist reactivity [27]Airway epithelial cells/mice Agonist reactivity [128]Pulmonary artery ECs/bovine Agonist reactivity [149]Lung epithelial cells/rat Northern blot [148]Type II epithelial cells/rat Agonist reactivity [151]1HAEo-, 16HBE14o-,and A549 /human

Northern blot [152]

Immunoblot [146]RTPCR, Northern blotAgonist reactivity [144]Confocal microscopy

Lung parenchyma/rabbit RTPCR [145]Pulmonary artery/rabbitPulmonary artery ECs/rabbit Airway smoothmuscle cells/mouse

Agonist reactivity [153]

Airway smooth musclecells/human

RTPCR [150]

31S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

events and ATP signaling remains under investigation. In sum,deviations from usual oxygen tensions, both high and low, cancause ATP release from endothelial and other lung cell types.

Another common pulmonary oxidant is ozone. It is animportant component of ambient air pollution and exposure toozone can cause exacerbation of asthma and other chronic lungdiseases [30–32]. Extracellular ATP protects lung epithelialcells against ozone toxicity [33]. P2Y receptor-mediatedincreases in ERK and Akt phosphorylations were importantsignaling events in this protection. Therefore, identification oflinks between ATP release and activation of survival pathwayswill illuminate a novel pathway in cellular defense againstoxidative stress. In this Review we highlight the unique role ofextracellular ATP as a danger signal, an “SOS” for host defenseduring pulmonary disorders caused by oxidative stress such asbronchopulmonary dysplasia and cystic fibrosis.

Purinoreceptor expression in pulmonary cells

Both P2Y and P2X receptors are ubiquitous in lung. Airwayepithelium has been one of the most heavily studied tissues foreffects of purinergic agonists [34–36]. Airway epithelial cellsrelease ATP and possess both P2X and P2Y receptors, as doendothelial cells [2,5,25,37]. Localization and distribution ofP2X receptor subtypes in lungs, along with methods ofdetection used, are summarized in Table 1. In general, themammalian pulmonary vasculature contains most P2X receptorsubtypes except for P2X6. P2X receptors are found in lungepithelium as well as endothelium. P2X4, P2X5, and P2X7 are

Table 1P2X receptor distribution in lungs

Receptorsubtype

Tissue/species Method of detection References

P2X Pulmonary vascular bed/cat Agonist reactivity [134]Pulmonary artery/human Agonist reactivity [135]Pulmonary artery/rat Agonist reactivity [136]

P2X1 Pulmonary artery ECs andLMVECs/human

RTPCR, Immunoblot [25]

Pulmonary artery ECs /human Northern blot [137]Pulmonary artery /rat Immunoblot, RTPCR,

in situ hybridization[138,139]

Intrapulmonary artery/pig Agonist reactivity [140]P2X2 Pulmonary artery ECs/rat RTPCR, in situ

hybridization[139]

P2X3 Pulmonary artery/human Northern blot, RTPCR [137]Pulmonary artery/rat Northern blot, RTPCR [138]

P2X4 Pulmonary artery/rat Immunochemistry [138]LMVECs/human Immunoblot, RTPCR [25]Bronchial epithelial cells/human Immunoblot [141]Pulmonary artery ECs/human Immunoblot [137]

P2X5 Pulmonary artery ECs/human Immunoblot [137]Bronchial epithelial cells/human Northern blot, RTPCR [141]LMVECs/human Immunoblot [25]

P2X6 Bronchial epithelial cells/human Immunoblot [141]P2X7 Pulmonary artery ECs/human Immunoblot [137,25]

Pulmonary artery/rat Immunochemistry [138]Lung/mice Immunoblot [142]Alveolar epithelial cells/rat RTPCR [143]

ECs, endothelial cells; LMVECs, lung microvascular endothelial cells.

P2Y4 Airway smooth musclecells/mouse

Agonist reactivity [153]

Airway smooth musclecells/human

RTPCR [150]

P2Y6 16 HBE14o-/human Confocal microscopy [144]1HAEo-, 16HBE14o-,and A549 /human

Northern blot, RTPCR [146]

P2Y11 16 HBE14o-/human Confocal microscopy [144]P2Y14 A549 BEAS-28/human Northern blot [154]

Agonist reactivity [146]

commonly present in bronchial and alveolar epithelial cells. Theapical and luminal surfaces contain one or more of thesereceptors that are often present as homomultimers andheteromultimers. Pulmonary artery endothelial cells containP2X1, P2X2, P2X4, P2X5, and P2X7 subtypes, whereas lungmicrovascular endothelial cells are reported to have P2X1,P2X4, P2X5, and P2X7 receptors.

P2Y receptors were identified in human lungs following thefinding of Knowles et al. [38] that extracellular nucleotidesstimulated chloride secretion in the lungs of cystic fibrosispatients. As for P2X receptors, the rank order potency ofagonists was initially the method of choice for defining thepresence of P2Y receptors and their subtypes (Table 2).Molecular cloning led to identification of several P2Y receptors,some of which are verified to be nucleotide receptors. P2Yreceptors have been identified and characterized in whole lungs,lung vasculature, parenchyma, smooth muscle cells, andendothelial and epithelial cells. P2Y receptors are present

32 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

along with P2X receptors, and a combination of their subtypesmay exist on the same cell. Novel P2Y receptors specific fordinucleotides have also been identified in human lung [39].

Endogenous ATP release in lung and its effects

Stimuli which cause ATP release by regulated mechanismsinclude mechanical stress/trauma, shear stress (increased bloodflow and or lung ventilation; volutrauma) [40], hypotonicmedium [41,42], inflammation, cAMP, and ATP itself [43]. Themechanisms responsible for the appearance of extracellular ATPare likely multiple and probably differ by cell type. First, it mayexit the cell via ATP-permeable release channels. Second, ATPmay be conducted down favorable concentration gradients bysuch channels, e.g., plasma membrane forms of the voltage-dependent anion channel (VDAC) [44], a mitochondrialchannel capable of conducting ATP and ADP. Third, adeninenucleotide transporters, such as the ABC (ATP-bindingcassette) family of transporters like the multidrug resistancechannel (MDR or Mrp) [45], may serve as transporters of ATPand many other substrates [46,47]. Another member of thisfamily, the cystic fibrosis transport regulator (CFTR), generallyis acknowledged to modulate extracellular transport of ATPthrough such pathways, but also is conceded not to be an ATPrelease channel itself [48]. ATP exchange with ADP, as inmitochondria, also is possible through ABC transporters.Fourth, ATP release may occur through vesicular exocytosis[49]. There is considerable support for such a mechanism ofrelease from various epithelia. Fifth, two or more such processescould act in combination. For example, ATP channels ortransporters could load ATP into vesicles which then fuse withthe plasma membrane and release their contents (exocytosis)(Fig. 2).

Fig. 2. Schematic representation of mechanisms of ATP release from cells. Upon stimand transporters. Cell death and cellular injury may also cause direct ATP release intobe metabolized to its degradation products (ADP, AMP, adenosine) by the action ofspecific receptors or be converted back to ATP by the reverse action of these enzym

Once released from the cell, extracellular adenine nucleotideconcentrations can be further dynamically controlled by (a)receptor interaction and/or uptake, and (b) cell surface enzymescalled ecto-enzymes. Hence, ATP can be broken down orconverted into lower energy forms, including ADP, AMP,cAMP, and adenosine. Ecto-apyrases break down ATP to ADPand 5′-AMP. Ecto-ATPases and ADPases also can act to breakdown ATP. Ecto-5′-nucleotidases convert 5′-AMP to adenosine[50,51]. These enzymes also can act in reverse to resynthesizeATP. In addition, more elaborate complexes may exist tosynthesize ATP extracellularly [52] (Fig. 2). Changes inexpression or activity of these ecto-enzymes can modifyphysiologic responses because the different metabolites gener-ated have maximal actions on different receptors. For example,the breakdown product adenosine triggers a host of physiologicactions through P1 receptors [53]. Adenosine may be ofparticular interest and importance in lung diseases like asthma[54–56] and cystic fibrosis [57]. Not only can ATP actindirectly via breakdown to adenosine, but, in addition, ATPcan upregulate or activate adenosine receptors to enhance theresponse [58,59].

The effect of ATP and its metabolites in the pulmonarycirculation is variable and depends on the stimulus, the effectormetabolite ,and the endpoint being studied. In the pulmonarycirculation ATP is released primarily from red blood cells. It isalso released by platelets and endothelial cells lining the vesselwalls. For example, collagen injection caused release of ADPand ATP from platelets in the coronary and pulmonarymicrocirculations and elsewhere [60]. Released ADP inducedplatelets to form aggregates that lodged in the vessels resultingin thrombocytopenia and cardiopulmonary dysfunction. In-creased adenosine and its degradation products in thepulmonary circulation are also observed upon anoxic exposure

ulation ATP is released either exocytotically or through transmembrane channelsthe extracellular milieu. Once released ATP may bind to the P2 receptor, or it canthe enzymes called ecto-nucleotidases. ADP, AMP, and adenosine may bind toes.

33S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

[61]. Efflux of [3H]purines from pulmonary artery induced bypotassium or calcium ionophore A23187 has also beeninvestigated [62]. Endogenous ATP released during increasesin flow rate dilates pulmonary vessels [63]. Mechanical stress-induced release of ATP also has been described in humanairway epithelial cells [64]. Recent studies indicate that ATPrelease following mechanical injury also promotes cellmigration, a critical event in epithelial wound closure, invarious epithelial cell types [65,66] including airway epithelialcells (A van der Vliet, personal communication). The role ofextracellular ATP and mechanical deformation-induced stimu-lation of alveolar type II cells to secrete surfactant has beencarefully explored [67–72]. ATP release and extracellular ATPalso can play a major modulatory role in histamine release fromhuman lung mast cells, and therefore, it may be mechanisticallyinvolved in human allergic/asthmatic reactions [73]. Thus, ATPrelease from one tissue/cell type can trigger a response inanother tissue/cell type, thereby establishing interaction.

Pulmonary epithelium is the primary barrier against varioustoxic chemical and mechanical environmental stresses includingoxidant gases and particulates. Preservation of airway andalveolar homeostasis is a necessary component of the host–defense function in lungs. ATP release upon stimulation of theluminal surface of the epithelium is known to cause enhancedfluid secretion and increased ciliary beat frequency which helpsclear the epithelial surface and restore alveolar homeostasis[74,75]. Extracellular ATP acts by triggering purinergicreceptors which, in turn, cause elevation of intracellular calciumconcentration [74]. Nucleotides within the airway surface liquid(ASL) also regulate airway epithelial ion transport rates by Ca2+-and protein kinase C-dependent mechanisms [57]. For thesereasons, a tremendous effort has been focused on developmentof purine analogs as therapeutic agents for treatment of cysticfibrosis. ATP signaling may regulate or modulate numerouscritical cell functions relevant to cystic fibrosis includingpromotion of: (1) volume homeostatic responses to alteredtonicity of the extracellular milieu [2,76,77], increased ciliarybeat frequency [4,78], epithelial cell secretion of ions [76,79–82], and (2) fluid [5,57,83], mucin secretion [6,84], and releaseof inflammatory cytokines [85–87]. Some studies suggest thatATP release from CF epithelial cells is deficient [37,88].However, others do not [89]. Studies regarding the potential ofCF airways to release ATP under basal conditions or in responseto stress appear to differ based on, among other things, thestressful stimulus. CF epithelial cells appear deficient in ATPrelease responses to osmotic stress, while they are reported tohave normal ATP release responses to shear. Braunstein et al.[77] recently reported that transformed non-CF human airwayepithelial cells and CF airway cells transfected with wild-typeCFTR could both undergo regulatory volume decrease (RVD) inresponse to hypotonic stress, and that these responses wereinhibited by blockade of purinergic signaling. By contrast, CFcell line IB3-1 or IB3-1 transfected with mutant CFTRs (DF508,G551D, or S1445X) could not produce RVDs. Nonetheless,IB3-1 cells could produce RVD if their P2Y G protein-coupledreceptors and/or P2X receptors were stimulated. This supportedprevious work by others that cell volume regulation is dependent

on extracellular ATP and is impaired in CF [37,48,90].Lazarowski et al. [57] recently reported that primary CF andcontrol, fully differentiated human bronchial epithelial cellsgrown on supports showed similar levels of apical ATP release,both at rest and following stimulation due to shear stress. Thereare numerous possibilities for different conclusions reached inthese two studies. The latter study utilized fully differentiatedprimary CF versus control airway epithelial cell cultures,whereas the former study used a CF cell line with its stablytransfected, CFTR-corrected control cell, as well as a number ofother transiently transfected CF cells bearing other CFTRmutants or empty vector. The study in primary cells focused onappearance of ATP and its breakdown products in theextracellular space in response to shear or under basalconditions, while the focus in the CF cell line and transfectionwas on RVD response to hypotonic stress and the potency ofvarious agonists/antagonists to correct or inhibit the deficient CFRVD responses. Nonetheless, in each study, CF cells differedfrom controls. In one, in RVD responses, and, in the other, in thatthey had lower levels of extracellular adenosine. Mechanicallyinduced ATP release and its effect in enhancing clearance ofsecretions from CF airways also have recently been described[91]. Taken together, these data indicate that cystic fibrosisairway epithelium, itself a target of profound oxidative stress,may also have altered signaling responses due to extracellularATP and/or adenosine.

P2 receptor knockouts in general are not embryonically lethaland survive with no apparent phenotypic abnormality. BecauseATP-dependent signaling is often more relevant to physiologicstress adaptation, it is conceivable that the important stressorsthat might cause difficulty for these various mice may not yethave been discovered. Moreover, the complexity of distributionand ligand-binding specificity of the P2 receptor family havemade it extremely difficult to assign a particular receptor tospecific physiological response. Knockout mice have, however,uncovered tissue-specific roles of P2Y receptor, as P2Y2−/−mice have decreased ATP-regulated chloride secretion intracheal epithelium [92]. P2Y4 knockout mice, on the otherhand, have complete loss of ATP-and UTP-induced chloridesecretory response in the jejunum as compared to the wild type[93,94]. Inhibition of platelet aggregation to ADP and increasedbleeding time in P2Y1 (−/−) and P2Y12 (−/−) mice have alsobeen reported [95]. Double knockout P2Y1 and P2Y2 (P2Y1/P2Y2−/−) mice have enhanced susceptibility to Pseudomonasaeruginosa-induced death, suggesting the involvement of thesereceptors in innate host defense of the lung [96]. P2X receptorknockout mice reveal important roles of these receptors in theneurogenic control of smooth muscle contraction, pain, boneformation, and inmacrophage function [97]. P2X3−/−, P2X2−/−,and P2X2/P2X3(Dbl−/−) mice present reduced urinary bladderreflexes and decreased pelvic afferent nerve activity in responseto bladder distension [98]. P2X2−/− and P2X2/P2X3(Dbl−/−)and P2X7−/− mice also exhibit reduced pain-related behaviors[98,99]. Genetic elimination of inotropic purinergic receptors(P2X2 and P2X3) eliminates taste responses in taste nerves[100]. In summary, a wide spectrum of physiologic abnormal-ities is revealed in P2 receptor knockout animals. Because of the

34 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

large numbers of receptors of both classes, P2Y and P2X, thelack of abnormal phenotype in many knockouts may be morerelated to redundancies in these systems than to overall lack ofimportance of function.

ATP-mediated protection against oxidative stress

As described above, in lungs, mechanical stress orintroduction of foreign particles on the luminal surface cantrigger nucleotide release. Subsequently, P2Y2 receptor-mediated secretion can be stimulated and larger amounts offluid generated, potentially contributing to airway particleclearance [101]. In addition to clearing the epithelial surfaceand restoring alveolar homeostasis, extracellular ATP alsocould protect pulmonary epithelium and endothelium againstoxidant stresses caused by exposure to hyperoxia and ozone[27,33]. We showed previously that hypoxic exposure protectslung endothelial cells against subsequent hyperoxic exposuretoxicity [21]. Hypoxic exposure, besides causing upregulationof important survival pathways, caused enhanced accumula-tion of ATP in the extracellular medium [22]. A similaraccumulation of ATP in extracellular medium was observed inhyperoxia-exposed cells. Removal of extracellular ATP orblockade of P2 receptors increased hyperoxia-induced toxicity[27]. This suggested that extracellular ATP and its signalingresponses are crucial for survival during hyperoxic exposures.Increased blood plasma ATP level in the pulmonarycirculation of enhanced oxygen-exposed fetal lambs also hasbeen demonstrated [102], indicating that these observations incell culture could be pertinent to in vivo exposure tohyperoxia.

Early enhancement of extracellular ATP content upon ozoneexposure also was observed in lung epithelial cells derived fromall levels of human and rat airways. Interestingly, ozone-mediated toxicity to lung epithelial cells was abolished bysupplemental extracellular ATP or stable ATP analog ATP-γ-S[33]. The inflammatory response of pulmonary endothelium cancause increased transendothelial permeability, contributing toextrusion of fluid and blood cells and resulting in lung edema.In addition to enhancing survival, extracellular ATP and itsanalogs enhance barrier function in pulmonary endothelial cells[103]. Barrier-protective properties of ATP have been describedin endothelial cells from other organs as well [104].Extracellular ATP stimulates airway epithelial cells to produceH2O2, a necessary substrate for the lactoperoxidase (LPO) anti-infection system, thus contributing to airway host defense [18].In another study, extracellular supplementation with ATP alsoprotected astrocytes against hydrogen peroxide-mediated celldeath [105]. Similarly, ATP supplementation may also protecthippocampal neurons from excitotoxic cell death caused byoverstimulation by glutamate [106]. Moreover, ATP releaseddue to brain injury mediates rapid recruitment of microglialcells, principal immune cells of brain, thus signaling neuronaltissue repair responses [107]. Cell death induced by hypoxia inrat cardiomyocyte was significantly reduced by extracellularUTP [108]. Extracellular ATP and adenosine are also known toplay complementary roles in ischemic preconditioning of heart

[109]. Administration of 2-methylthio-ATP, an ATP analog, inmice protected them against subsequent endotoxin-induceddeath [110]. Thus, ATP signaling can mediate a variety ofprotective responses relevant to oxidative stress in lung andother organs.

ATP is also released in response to other oxidants likehydrogen peroxide [111], and this may be pertinent inprotection of cells against hydrogen peroxide-induced death[112]. Extracellular ATP as well as 2-methylthio-ATP and ATP-γ-S (P2Y purinoceptor agonists) can inhibit ischemia-inducedlactate dehydrogenase (LDH) release [113]. Thus, it appearsthat endogenously released ATP or exogenously added ATP canprotect cells against a variety of oxidants.

In addition to ATP mediating tissue protection through its P2receptors, downstream metabolites of ATP like adenosine alsocan protect against oxidants. Adenosine also has beenimplicated in protection of pulmonary endothelial cells againstH2O2-mediated cytotoxicity [114]. Recently, A2A adenosinereceptors were found to be important in limiting andterminating both tissue-specific and systemic inflammatoryresponses [115,116]. Thus, adenosine and ATP may beimportant autocrine/paracrine regulators mediating cellularprotection and regeneration after ischemia [109,117], and P1and P2 receptors may cooperate to limit tissue damage due tooxidants.

ATP analogs and P2Y2 receptor agonists are a new class ofcompounds being developed for the treatment of CF lungdisease. A number of P2Y2 agonist compounds have beenevaluated in healthy subjects and patients with CF. Mostrecently, INS37217, a metabolically stable and potent P2Y2

agonist has been developed, and studies have shown it to bewell-tolerated when given via inhalation. This compound iscurrently being evaluated in children and adults with CF lungdisease [118]. Results on preliminary trials with oral andintravenously administered ATP, as an analgesic, also areencouraging for the treatment of patients with postherpeticneuralgia and low back pain [119,120]. ATP release andsubsequent signaling via P2X receptors mediate excitation ofsensory neurons and evoke muscle contraction in humanurinary bladder. Functional defects in the P2X receptorsignaling are associated with a variety of urologic diseases[121]. Studies on ATP-mediated signaling present a potentialapplication in relation to the diagnosis and treatment of urinarydysfunction. ATP may also inhibit pathological renal cystgrowth through P2X7 signaling [122]. Because of potentialcardiovascular effects of extracellular ATP itself, analogstargeted to one or more receptor(s) may offer greater specificityof effect(s) in therapy.

Signaling events associated with ATP-mediated protection

P2X receptors mediate “rapid: nonselective passage ofcations (Na+, K+, Ca2+) across cell membranes resulting inincreased intracellular Ca2+ and depolarization [123,124].Conductance of extracellular Ca2+ through such channels is asignificant source of increased intracellular Ca2+. Suchresponses are very rapid and do not involve production of

35S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

second messengers. These signals are important for rapidneuronal signaling and regulation of muscle contractility. P2Yreceptors are G-protein-coupled receptors and most of them actvia G-protein coupling to activate phospholipase C (PLC),leading to formation of inositol triphosphate (IP3) andmobilization of intracellular Ca2+. Coupling to adenylatecyclase by some P2Y receptors (e.g., P2Y12, P2Y13, andP2Y14) has also been described [125]. The response time ofP2Y receptors is longer than the rapid responses mediated byP2X receptors, because P2Y-mediated responses involveformation of second messengers. IP3 formation and Ca2+

mobilization can stimulate a variety of signaling pathwaysincluding protein kinase C (PKC), phospholipase A2 (PLA2),etc. PKC, in turn, may activate PLC, PLD, and MAP kinasepathways [126]. These responses, as well as secondaryformation of various eicosanoids, could be important insignaling P2Y-mediated tissue protective responses.

Extracellular ATP can mediate increases in intracellularcalcium via P2Y receptors in alveolar type II epithelial cells,bronchial epithelial cells, and lung fibroblasts [127–129]. Ourunderstanding of ATP-mediated signaling events and theirmodulation following oxidative stress are beginning to unfold.A better understanding of these processes could be helpful indevising protection strategies.

Fig. 3. Survival signaling responses activated by oxidant gases via extracellular ATPby niflumic acid (chloride channel inhibitor), colchicine (a microtubule disrupting agePI3K) suggested involvement of a complex mechanism in hyperoxia-induced ATPU0126 (a MEK specific inhibitor), Y27637 (a rho kinase inhibitor) brefeldin A (a veindicating calcium-dependent vesicular exocytosis as the primary mechanism. ExtraMEK. Activation of PI3K and MEK was accompanied by increased activity of theirfor PI3K, and ERK 1/2 for MEK). Action of apyrase (an ATP scavenger), suramin (signaling and caused increased cell death in both hyperoxia and ozone. Extracellularlysignaling pathways and their metabolic effects.

Oxidative stress due to hyperoxic exposure caused accumu-lation of ATP in the extracellular milieu of HLMVECs by aPI3K-dependent mechanism. ATP release could be modulated inpart by inhibition of CFTR or of microtubular function.Scavenging of extracellular ATP before and during hyperoxicexposure caused enhanced toxicity, indicating the significanceof extracellular nucleotide for cellular protection. Similarly,exposure to lung epithelial-like A549 cells to ozone causedincreases in extracellular ATP (Fig. 3). Therein, a complex-regulated mechanism involving vesicular exocytosis wasobserved for ATP release. Here too, we found that extracellularATP was indispensable for survival in ozone. Protective effectsof extracellular ATP against hyperoxia-and ozone-mediatedtoxicity were due to two key pathways: extracellular signal-regulated protein kinase (ERK) and PI3K. These protein kinasesare critical regulators of cell survival and proliferation[130,131]. Extracellular ATP induced activation of ERK,PI3K, and mTOR (mammalian target of rapamycin), primarilythrough activation of P2Y2 and P2Y6 receptors in HLMVECs(Fig. 3). mTOR mediates growth and survival by regulatingcellular metabolism. Rapamycin (a triene macrolide antibiotic)binds to a receptor protein (FKBP12), and the rapamycin/FKBP12 complex then binds to mTOR and prevents interactionof mTOR with target proteins [132]. Rapamycin also caused

release. Cellular exposure to ozone and hyperoxia causes ATP release. Inhibitionnt), LY 294002 (a specific PI3K inhibitor), and wortmannin (another inhibitor ofrelease. Ozone-mediated ATP release was attenuated by colchicine, LY294002,sicular transport inhibitor), and BAPTA-AM (an intracellular calcium chelator),cellular ATP acted via P2Y receptors to activate survival kinases like PI3K anddownstream target protein kinases (Akt or protein kinase B, mTOR and p70S6KP2 receptor inhibitor), and Cibacron Blue (P2Y receptor inhibitor) blocked thisreleased ATP signals to prevent oxidant gas-induced death via Akt and /or ERK

36 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

enhanced cell death in hyperoxia, further supporting acontributing role for mTOR. Moreover, enhanced P2Y recep-tor-mediated glucose uptake, also inhibited by rapamycin,provided additional evidence for a protective role for mTOR.ERK activation due to ATP release in hyperoxia likelycontributed to enhanced survival. ATP release increasedarachidonic acid release and enhanced prostaglandin (PGE2)synthesis, which may also have contributed to cellularadaptation and resistance to oxidative damage. Activation ofERK and Akt was also critical for survival during ozoneexposure. Activation of these kinases was linked to increasedglucose metabolism, since ATP supplementation enhancedglucose uptake during ozone exposure. These studies demon-strate a novel survival-enhancing function for ATP release andextracellular ATP in the response of cells against oxidativestress. Despite evidence that stimulus-induced, regulated ATPrelease can inhibit oxidative damage, unregulated or unrestrictedATP release in severe tissue injury can exacerbate or enhancetissue damage [133].

Activation of nucleotide receptors in pulmonary endothelialand epithelial cells is coupled to ERK and PI3K pathways thatregulate survival during oxidant stress. Deficits in these signalsdue to impaired nucleotide production or release couldcontribute to disease pathogenesis or exacerbate existingconditions. Because ATP analogs already are in clinical trialsfor certain lung diseases, these findings could have applicationin prevention or treatment of respiratory disorders such asasthma, BPD, and air pollution-related morbidity in vulnerablepopulations. An improved understanding of ATP release,extracellular ATP metabolism, and relevant downstreamsignaling mechanisms also could lead to the development ofnovel therapeutic agents.

Acknowledgments

This publication was made possible by Grant ES014448-01from the National Institute of Environmental Health Sciences(NIEHS), NIH. The authors are also grateful to GabrieleCheatham for the preparation of the manuscript.

References

[1] Bodin, P.; Burnstock, G. Purinergic signalling: ATP release. Neurochem.Res. 26:959–969; 2001.

[2] Schwiebert, E. M.; Zsembery, A. Extracellular ATP as a signalingmolecule for epithelial cells. Biochim. Biophys. Acta 1615:7–32; 2003.

[3] Burnstock, G. Purinergic signalling. Br. J. Pharmacol. 147(Suppl. 1):S172–S181; 2006.

[4] Ostrom, R. S.; Gregorian, C.; Insel, P. A. Cellular release of and responseto ATP as key determinants of the set-point of signal transductionpathways. J. Biol. Chem. 275:11735–11739; 2000.

[5] Novak, I. ATP as a signaling molecule: the exocrine focus. News Physiol.Sci. 18:12–17; 2003.

[6] Dawicki, D. D.; Chatterjee, D.; Wyche, J.; Rounds, S. Extracellular ATPand adenosine cause apoptosis of pulmonary artery endothelial cells. Am.J. Physiol. 273:L485–L494; 1997.

[7] Mason, H. S.; Bourke, S.; Kemp, P. J. Selective modulation of ligand-gated P2X purinoceptor channels by acute hypoxia is mediated byreactive oxygen species. Mol. Pharmacol. 66:1525–1535; 2004.

[8] Warburton, D.; Buckley, S.; Cosico, L.; Forman, H. J. Sublethal oxidant

injury inhibits signal transduction in rat type II pneumocytes. Am. J.Physiol. 257:L217–L220; 1989.

[9] Maggirwar, S.; Dhanraj, D.; Somani, S.; Ramkumar, V. Adenosine acts asan endogenous activator of the celluar antioxidant defense system.Biochem. Biophys. Res. Commun. 201:508–515; 1994.

[10] Hirai, K.; Ashraf, M. Modulation of adenosine effects in attenuation ofischemia and reperfusion injury in rat heart. J. Mol. Cell. Cardiol.30:1803–1815; 1998.

[11] Kitakaze, M.; Minamino, T.; Node, K.; Takashima, S.; Funaya, H.;Kuzuya, T.; Hori, M. Adenosine and cardioprotection in the diseasedheart. Jpn. Circ. J. 63:231–243; 1999.

[12] Cross, H. R.; Murphy, E.; Black, R. G.; Auchampach, J.; Steenbergen, C.Overexpression of A(3) adenosine receptors decreases heart rate,preserves energetics, and protects ischemic hearts. Am. J. Physiol.Heart Circ. Physiol. 283:H1562–H1568; 2002.

[13] Maddock, H. L.; Mocanu, M. M.; Yellon, D. M. Adenosine A(3) receptoractivation protects the myocardium from reperfusion/reoxygenationinjury. Am. J. Physiol. Heart Circ. Physiol. 283:H1307–H1313; 2002.

[14] Aalto, T. K.; Raivio, K. O. Adenine nucleotide depletion from endothelialcells exposed to xanthine oxidase.Am. J. Physiol. 259:C883–C888; 1990.

[15] Aalto, T. K.; Raivio, K. O. Mechanisms of adenine nucleotide depletionfrom endothelial cells exposed to reactive oxygen metabolites. FreeRadic. Biol. Med. 14:177–183; 1993.

[16] Cheng, Y.; Chen, M.; James-Kracke, M.; Wixom, P.; Sun, A. Y. Enhancedlipid peroxidation by extracellular ATP in PC12 cells. Neurochem. Res.21:27–33; 1996.

[17] Wilkinson, J. A.; Jacob, R. Agonist-induced calcium and oxidative stressresponses in endothelial cells. Biochem. Soc. Trans. 31:960–962; 2003.

[18] Forteza, R.; Salathe, M.; Miot, F.; Forteza, R.; Conner, G. E. Regulatedhydrogen peroxide production by Duox in human airway epithelial cells.Am. J. Respir. Cell Mol. Biol. 32:462–469; 2005.

[19] Judkins, C. P.; Sobey, C. G.; Dang, T. T.; Miller, A. A.; Dusting, G. J.;Drummond, G. R. NADPH-induced contractions of mouse aorta do notinvolve NADPH oxidase: a role for P2X receptors. J. Pharmacol. Exp.Ther. 2006 [Electronic publication ahead of print].

[20] Crapo, J. D.; Barry, B. E.; Foscue, H. A.; Shelburne, J. Structural andbiochemical changes in rat lungs occurring during exposures to lethal andadaptive doses of oxygen. Am. Rev. Respir. Dis. 122:123–143; 1980.

[21] Ahmad, S.; Ahmad, A.; Gerasimovskaya, E.; Stenmark, K. R.; Allen,C. B.; White, C. W. Hypoxia protects human lung microvascularendothelial and epithelial-like cells against oxygen toxicity: role ofphosphatidylinositol 3-kinase. Am. J. Respir. Cell Mol. Biol. 28:179–187; 2003.

[22] Gerasimovskaya, E. V.; Ahmad, S.; White, C. W.; Jones, P. L.; Carpenter,T. C.; Stenmark, K. R. Extracellular ATP is an autocrine/paracrineregulator of hypoxia-induced adventitial fibroblast growth. Signalingthrough extracellular signal-regulated kinase-1/2 and the Egr-1 transcrip-tion factor. J. Biol. Chem. 277:44638–44650; 2002.

[23] Gerasimovskaya, E. V.; Davie, N. J.; Ahmad, S.; Tucker, D. A.; White,C. W.; Stenmark, K. R. Extracellular adenosine triphosphate: a potentialregulator of vasa vasorum neovascularization in hypoxia-inducedpulmonary vascular remodeling. Chest 128:608S–610S; 2005.

[24] Dubyak, G. R. Focus on “extracellular ATP signaling and P2X nucleotidereceptors in monolayers of primary human vascular endothelial cells”.Am. J. Physiol., Cell Physiol. 282:C242–C244; 2002.

[25] Schwiebert, L. M.; Rice, W. C.; Kudlow, B. A.; Taylor, A. L.; Schwiebert,E. M. Extracellular ATP signaling and P2X nucleotide receptors inmonolayers of primary human vascular endothelial cells. Am. J. Physiol.,Cell Physiol. 282:C289–C301; 2002.

[26] Yamakawa, M.; Liu, L. X.; Date, T.; Belanger, A. J.; Vincent, K. A.;Akita, G. Y.; Kuriyama, T.; Cheng, S. H.; Gregory, R. J.; Jiang, C.Hypoxia-inducible factor-1 mediates activation of cultured vascularendothelial cells by inducing multiple angiogenic factors. Circ. Res.93:664–673; 2003.

[27] Ahmad, S.; Ahmad, A.; Ghosh, M.; Leslie, C. C.; White, C. W.Extracellular ATP-mediated signaling for survival in hyperoxia-inducedoxidative stress. J. Biol. Chem. 279:16317–16325; 2004.

[28] Allen, C. B.; Guo, X. L.; White, C. W. Changes in pulmonary expression

37S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

of hexokinase and glucose transporter mRNAs in rats adapted tohyperoxia. Am. J. Physiol. 274:L320–L329; 1998.

[29] Ahmad, A.; Ahmad, S.; Schneider, B. K.; Allen, C. B.; Chang, L. Y.;White, C. W. Elevated expression of hexokinase II protects human lungepithelial-like A549 cells against oxidative injury. Am. J. Physiol., LungCell Mol. Physiol. 283:L573–L584; 2002.

[30] Thurston, G. D.; Bates, D. V. Air pollution as an underappreciated causeof asthma symptoms. JAMA 290:1915–1917; 2003.

[31] Goss, C. H.; Newsom, S. A.; Schildcrout, J. S.; Sheppard, L.; Kaufman,J. D. Effect of ambient air pollution on pulmonary exacerbations andlung function in cystic fibrosis. Am. J. Respir. Crit. Care Med.169:816–821; 2004.

[32] Mudway, I. S.; Kelly, F. J. An investigation of inhaled ozone dose and themagnitude of airway inflammation in healthy adults. Am. J. Respir. Crit.Care Med. 169:1089–1095; 2004.

[33] Ahmad, S.; Ahmad, A.; McConville, G.; Schneider, B. K.; Allen, C. B.;Manzer, R.; Mason, R. J.; White, C. W. Lung epithelial cells release ATPduring ozone exposure: signaling for cell survival. Free Radic. Biol. Med.39:213–226; 2005.

[34] Davis, C. W. Regulation of mucin secretion from in vitro cellular models.Novartis Found. Symp. 248:113–125 (discussion 125-31, 277-82); 2002.

[35] Kunzelmann, K.; Mall, M. Pharmacotherapy of the ion transport defect incystic fibrosis: role of purinergic receptor agonists and other potentialtherapeutics. Am. J. Respir. Med. 2:299–309; 2003.

[36] Hayashi, T.; Kawakami, M.; Sasaki, S.; Katsumata, T.; Mori, H.; Yoshida,H.; Nakahari, T. ATP regulation of ciliary beat frequency in rat trachealand distal airway epithelium. Exp. Physiol. 90:535–544; 2005.

[37] Taylor, A. L.; Kudlow, B. A.; Marrs, K. L.; Gruenert, D. C.; Guggino,W. B.; Schwiebert, E. M. Bioluminescence detection of ATP releasemechanisms in epithelia. Am. J. Physiol. 275:C1391–C1406; 1998.

[38] Knowles, M. R.; Clarke, L. L.; Boucher, R. C. Activation by extracellularnucleotides of chloride secretion in the airway epithelia of patients withcystic fibrosis. N. Engl. J. Med. 325:533–538; 1991.

[39] Laubinger, W.; Wang, H.; Welte, T.; Reiser, G. P2Y receptor specific fordiadenosine tetraphosphate in lung: selective inhibition by suramin,PPADS, Ip5I, and not by MRS-2197. Eur. J. Pharmacol. 468:9–14;2003.

[40] Rich, P. B.; Douillet, C. D.; Mahler, S. A.; Husain, S. A.; Boucher,R. C. Adenosine triphosphate is released during injurious mechanicalventilation and contributes to lung edema. J. Trauma 55:290–297;2003.

[41] Wang, Y.; Roman, R.; Lidofsky, S. D.; Fitz, J. G. Autocrine signalingthrough ATP release represents a novel mechanism for cell volumeregulation. Proc. Natl. Acad. Sci. USA 93:12020–12025; 1996.

[42] Feranchak, A. P.; Roman, R. M.; Schwiebert, E. M.; Fitz, J. G.Phosphatidylinositol 3-kinase contributes to cell volume regulationthrough effects on ATP release. J. Biol. Chem. 273:14906–14911; 1998.

[43] Anderson, C. M.; Bergher, J. P.; Swanson, R. A. ATP-induced ATPrelease from astrocytes. J. Neurochem. 88:246–256; 2004.

[44] Okada, S. F.; O'Neal, W. K.; Huang, P.; Nicholas, R. A.; Ostrowski, L. E.;Craigen, W. J.; Lazarowski, E. R.; Boucher, R. C. Voltage-dependentanion channel-1 (VDAC-1) contributes to ATP release and cell volumeregulation in murine cells. J. Gen. Physiol. 124:513–526; 2004.

[45] Roman, R. M.; Lomri, N.; Braunstein, G.; Feranchak, A. P.; Simeoni,L. A.; Davison, A. K.; Mechetner, E.; Schwiebert, E. M.; Fitz, J. G.Evidence for multidrug resistance-1 P-glycoprotein-dependent regula-tion of cellular ATP permeability. J. Membr. Biol. 183:165–173;2001.

[46] Schwiebert, E. M. ABC transporter-facilitated ATP conductive transport.Am. J. Physiol. 276:C1–C8; 1999.

[47] Roman, R. M.; Wang, Y.; Lidofsky, S. D.; Feranchak, A. P.; Lomri, N.;Scharschmidt, B. F.; Fitz, J. G. Hepatocellular ATP-binding cassetteprotein expression enhances ATP release and autocrine regulation of cellvolume. J. Biol. Chem. 272:21970–21976; 1997.

[48] Braunstein, G. M.; Roman, R. M.; Clancy, J. P.; Kudlow, B. A.; Taylor,A. L.; Shylonsky, V. G.; Jovov, B.; Peter, K.; Jilling, T.; Ismailov, I. I.;Benos, D. J.; Schwiebert, L. M.; Fitz, J. G.; Schwiebert, E. M. Cysticfibrosis transmembrane conductance regulator facilitates ATP release by

stimulating a separate ATP release channel for autocrine control of cellvolume regulation. J. Biol. Chem. 276:6621–6630; 2001.

[49] Gatof, D.; Kilic, G.; Fitz, J. G. Vesicular exocytosis contributes tovolume-sensitive ATP release in biliary cells. Am. J. Physiol.: Gastro-intest. Liver Physiol. 286:G538–G546; 2004.

[50] Joseph, S. M.; Buchakjian, M. R.; Dubyak, G. R. Colocalization of ATPrelease sites and ecto-ATPase activity at the extracellular surface ofhuman astrocytes. J. Biol. Chem. 278:23331–23342; 2003.

[51] Joseph, S. M.; Pifer, M. A.; Przybylski, R. J.; Dubyak, G. R. MethyleneATP analogs as modulators of extracellular ATP metabolism andaccumulation. Br. J. Pharmacol. 142:1002–1014; 2004.

[52] Kim, B. W.; Choo, H. J.; Lee, J. W.; Kim, J. H.; Ko, Y. G. ExtracellularATP is generated by ATP synthase complex in adipocyte lipid rafts. Exp.Mol. Med. 36:476–485; 2004.

[53] Bucheimer, R. E.; Linden, J. Purinergic regulation of epithelial transport.J. Physiol. 555:311–321; 2004.

[54] Rorke, S.; Holgate, S. T. Targeting adenosine receptors: novel therapeutictargets in asthma and chronic obstructive pulmonary disease. Am. J.Respir. Med. 1:99–105; 2002.

[55] Ryzhov, S.; Goldstein, A. E.; Matafonov, A.; Zeng, D.; Biaggioni, I.;Feoktistov, I. Adenosine-activated mast cells induce IgE synthesis by Blymphocytes: an A2B-mediated process involving Th2 cytokines IL-4and IL-13 with implications for asthma. J. Immunol. 172:7726–7733;2004.

[56] Sun, C. X.; Young, H. W.; Molina, J. G.; Volmer, J. B.; Schnermann,J.; Blackburn, M. R. A protective role for the A1 adenosine receptorin adenosine-dependent pulmonary injury. J. Clin. Invest. 115:35–43;2005.

[57] Lazarowski, E. R.; Tarran, R.; Grubb, B. R.; van Heusden, C. A.; Okada,S.; Boucher, R. C. Nucleotide release provides a mechanism for airwaysurface liquid homeostasis. J. Biol. Chem. 279:36855–36864; 2004.

[58] Almeida, T.; Rodrigues, R. J.; de Mendonca, A.; Ribeiro, J. A.; Cunha, R. A.Purinergic P2 receptors trigger adenosine release leading to adenosine A2Areceptor activation and facilitation of long-term potentiation in rathippocampal slices. Neuroscience 122:111–121; 2003.

[59] Matsuoka, I.; Ohkubo, S. ATP-and adenosine-mediated signaling inthe central nervous system: adenosine receptor activation by ATPthrough rapid and localized generation of adenosine by ecto-nucleotidases. J. Pharmacol. Sci. 94:95–99; 2004.

[60] Kobayashi, I.; Didisheim, P. Cardiopulmonary dysfunctions caused byintravenous collagen-induced release of ADP and ATP from platelets.Jpn. Circ. J. 39:1205–1214; 1975.

[61] Mentzer R. M., Jr.; Rubio, R.; Berne, R. M. Release of adenosine byhypoxic canine lung tissue and its possible role in pulmonary circulation.Am. J. Physiol. 229:1625–1631; 1975.

[62] Katsuragi, T.; Su, C. Purine release from vascular adrenergic nerves byhigh potassium and a calcium ionophore, A-23187. J. Pharmacol. Exp.Ther. 215:685–690; 1980.

[63] Hassessian, H.; Bodin, P.; Burnstock, G. Blockade by glibenclamide ofthe flow-evoked endothelial release of ATP that contributes tovasodilatation in the pulmonary vascular bed of the rat. Br. J. Pharmacol.109:466–472; 1993.

[64] Grygorczyk, R.; Hanrahan, J. W. CFTR-independent ATP release fromepithelial cells triggered by mechanical stimuli. Am. J. Physiol. 272:C1058–C1066; 1997.

[65] Dignass, A. U.; Becker, A.; Spiegler, S.; Goebell, H. Adenine nucleotidesmodulate epithelial wound healing in vitro. Eur. J. Clin. Invest.28:554–561; 1998.

[66] Klepeis, V. E.; Weinger, I.; Kaczmarek, E.; Trinkaus-Randall, V. P2Yreceptors play a critical role in epithelial cell communication andmigration. J. Cell Biochem. 93:1115–1133; 2004.

[67] Dietl, P.; Haller, T.; Mair, N.; Frick, M. Mechanisms of surfactantexocytosis in alveolar type II cells in vitro and in vivo. News Physiol. Sci.16:239–243; 2001.

[68] Gilfillan, A. M.; Rooney, S. A. Functional evidence for involvement ofP2 purinoceptors in the ATP stimulation of phosphatidylcholine secretionin type II alveolar epithelial cells. Biochim. Biophys. Acta 959:31–37;1988.

38 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

[69] Mason, R. J.; Voelker, D. R. Regulatory mechanisms of surfactantsecretion. Biochim. Biophys. Acta 1408:226–240; 1998.

[70] Rice, W. R.; Singleton, F. M. P2-purinoceptors regulate surfactantsecretion from rat isolated alveolar type II cells. Br. J. Pharmacol.89:485–491; 1986.

[71] Patel, A. S.; Reigada, D.; Mitchell, C. H.; Bates, S. R.; Margulies, S. S.;Koval, M. Paracrine stimulation of surfactant secretion by extracellularATP in response to mechanical deformation. Am. J. Physiol., Lung CellMol. Physiol. 289:L489–L496; 2005.

[72] Wirtz, H. R.; Dobbs, L. G. Calcium mobilization and exocytosis after onemechanical stretch of lung epithelial cells. Science 250:1266–1269;1990.

[73] Schulman, E. S.; Glaum, M. C.; Post, T.; Wang, Y.; Raible, D. G.;Mohanty, J.; Butterfield, J. H.; Pelleg, A. ATP modulates anti-IgE-induced release of histamine from human lung mast cells. Am. J. Respir.Cell Mol. Biol. 20:530–537; 1999.

[74] Korngreen, A.; Priel, Z. Purinergic stimulation of rabbit ciliatedairway epithelia: control by multiple calcium sources. J. Physiol. 497(Pt 1):53–66; 1996.

[75] Homolya, L.; Steinberg, T. H.; Boucher, R. C. Cell to cell communicationin response to mechanical stress via bilateral release of ATP and UTP inpolarized epithelia. J. Cell Biol. 150:1349–1360; 2000.

[76] Hwang, T. H.; Schwiebert, E. M.; Guggino, W. B. Apical and basolateralATP stimulates tracheal epithelial chloride secretion via multiplepurinergic receptors. Am. J. Physiol. 270:C1611–C1623; 1996.

[77] Braunstein, G. M.; Zsembery, A.; Tucker, T. A.; Schwiebert, E. M.Purinergic signaling underlies CFTR control of human airway epithelialcell volume. J. Cyst. Fibros. 3:99–117; 2004.

[78] Barrera, N. P.; Morales, B.; Villalon, M. Plasma and intracellularmembrane inositol 1,4,5-trisphosphate receptors mediate the Ca(2+)increase associated with the ATP-induced increase in ciliary beatfrequency. Am. J. Physiol. Cell Physiol. 287:C1114–C1124; 2004.

[79] Stutts, M. J.; Fitz, J. G.; Paradiso, A. M.; Boucher, R. C. Multiple modesof regulation of airway epithelial chloride secretion by extracellular ATP.Am. J. Physiol. 267:C1442–C1451; 1994.

[80] Schwiebert, E. M.; Egan, M. E.; Hwang, T. H.; Fulmer, S. B.; Allen, S. S.;Cutting, G. R.; Guggino, W. B. CFTR regulates outwardly rectifyingchloride channels through an autocrine mechanism involving ATP. Cell81:1063–1073; 1995.

[81] McGill, J. M.; Basavappa, S.; Mangel, A. W.; Shimokura, G. H.;Middleton, J. P.; Fitz, J. G. Adenosine triphosphate activates ionpermeabilities in biliary epithelial cells. Gastroenterology 107:236–243;1994.

[82] Roman, R. M.; Feranchak, A. P.; Davison, A. K.; Schwiebert, E. M.;Fitz, J. G. Evidence for Gd(3+) inhibition of membrane ATPpermeability and purinergic signaling. Am. J. Physiol. 277:G1222–G1230; 1999.

[83] Schlenker, T.; Romac, J. M.; Sharara, A. I.; Roman, R. M.; Kim, S. J.;LaRusso, N.; Liddle, R. A.; Fitz, J. G. Regulation of biliary secretionthrough apical purinergic receptors in cultured rat cholangiocytes. Am. J.Physiol. 273:G1108–G1117; 1997.

[84] Kim, K. C.; Lee, B. C. P2 purinoceptor regulation of mucin release byairway goblet cells in primary culture. Br. J. Pharmacol. 103:1053–1056;1991.

[85] Verhoef, P. A.; Estacion, M.; Schilling, W.; Dubyak, G. R. P2X7 receptor-dependent blebbing and the activation of Rho-effector kinases, caspases,and IL-1 beta release. J. Immunol. 170:5728–5738; 2003.

[86] Gudipaty, L.; Humphreys, B. D.; Buell, G.; Dubyak, G. R. Regulation ofP2X(7) nucleotide receptor function in human monocytes by extracellularions and receptor density. Am. J. Physiol., Cell Physiol. 280:C943–C953;2001.

[87] Suzuki, T.; Hide, I.; Ido, K.; Kohsaka, S.; Inoue, K.; Nakata, Y.Production and release of neuroprotective tumor necrosis factor by P2X7receptor-activated microglia. J. Neurosci. 24:1–7; 2004.

[88] Naumann, N.; Siratska, O.; Gahr, M.; Rosen-Wolff, A. P-glycopro-tein expression increases ATP release in respiratory cystic fibrosiscells. J. Cyst. Fibros. 4:157–168; 2005.

[89] Donaldson, S. H.; Lazarowski, E. R.; Picher, M.; Knowles, M. R.; Stutts,

M. J.; Boucher, R. C. Basal nucleotide levels, release, and metabolism innormal and cystic fibrosis airways. Mol. Med. 6:969–982; 2000.

[90] Valverde, M. A.; Bond, T. D.; Hardy, S. P.; Taylor, J. C.; Higgins, C. F.;Altamirano, J.; Alvarez-Leefmans, F. J. The multidrug resistance P-glycoprotein modulates cell regulatory volume decrease. EMBO J.15:4460–4468; 1996.

[91] Tarran, R.; Button, B.; Picher, M.; Paradiso, A. M.; Ribeiro, C. M.;Lazarowski, E. R.; Zhang, L.; Collins, P. L.; Pickles, R. J.; Fredburg, J. J.;Boucher, R. C. Normal and cystic fbrosis airway surface liquidhomeostasis: The effects of phasic shear stress and viral infections.J. Biol. Chem. 280:35751–35759; 2005.

[92] Cressman, V. L.; Lazarowski, E.; Homolya, L.; Boucher, R. C.; Koller,B. H.; Grubb, B. R. Effect of loss of P2Y(2) receptor gene expressionon nucleotide regulation of murine epithelial Cl(-) transport. J. Biol.Chem. 274:26461–26468; 1999.

[93] Dubyak, G. R. Knock-out mice reveal tissue-specific roles of P2Yreceptor subtypes in different epithelia. Mol. Pharmacol. 63:773–776;2003.

[94] Robaye, B.; Ghanem, E.; Wilkin, F.; Fokan, D.; Van Driessche, W.;Schurmans, S.; Boeynaems, J. M.; Beauwens, R. Loss of nucleotideregulation of epithelial chloride transport in the jejunum of P2Y4-nullmice. Mol. Pharmacol. 63:777–783; 2003.

[95] Leon, C.; Freund, M.; Ravanat, C.; Baurand, A.; Cazenave, J. P.; Gachet,C. Key role of the P2Y(1) receptor in tissue factor-induced thrombin-dependent acute thromboembolism: studies in P2Y(1)-knockout mice andmice treated with a P2Y(1) antagonist. Circulation 103:718–723; 2001.

[96] Geary, C.; Akinbi, H.; Korfhagen, T.; Fabre, J. E.; Boucher, R.; Rice, W.Increased susceptibility of purinergic receptor-deficient mice to lunginfection with Pseudomonas aeruginosa. Am. J. Physiol., Lung Cell Mol.Physiol. 289:L890–L895; 2005.

[97] Boeynaems, J. M.; Communi, D.; Gonzalez, N. S.; Robaye, B. Overviewof the P2 receptors. Semin. Thromb. Hemost. 31:139–149; 2005.

[98] Cockayne, D. A.; Dunn, P. M.; Zhong, Y.; Rong, W.; Hamilton, S. G.;Knight, G. E.; Ruan, H. Z.; Ma, B.; Yip, P.; Nunn, P.; McMahon, S. B.;Burnstock, G.; Ford, A. P. P2X2 knockout mice and P2X2/P2X3 doubleknockout mice reveal a role for the P2X2 receptor subunit in mediatingmultiple sensory effects of ATP. J. Physiol. 567:621–639; 2005.

[99] Chessell, I. P.; Hatcher, J. P.; Bountra, C.; Michel, A. D.; Hughes, J. P.;Green, P.; Egerton, J.; Murfin, M.; Richardson, J.; Peck, W. L.; Grahames,C. B.; Casula, M. A.; Yiangou, Y.; Birch, R.; Anand, P.; Buell, G. N.Disruption of the P2X7 purinoceptor gene abolishes chronic inflamma-tory and neuropathic pain. Pain 114:386–396; 2005.

[100] Finger, T. E.; Danilova, V.; Barrows, J.; Bartel, D. L.; Vigers, A. J.; Stone,L.; Hellekant, G.; Kinnamon, S. C. ATP signaling is crucial forcommunication from taste buds to gustatory nerves. Science 310:1495–1499; 2005.

[101] Leipziger, J. Control of epithelial transport via luminal P2 receptors. Am.J. Physiol. Renal Physiol. 284:F419–F432; 2003.

[102] Konduri, G. G.; Mattei, J. Role of oxidative phosphorylation and ATPrelease in mediating birth-related pulmonary vasodilation in fetal lambs.Am. J. Physiol, Heart Circ. Physiol. 283:H1600–H1608; 2002.

[103] Kolosova, I. A.; Mirzapoiazova, T.; Adyshev, D.; Usatyuk, P.; Romer,L. H.; Jacobson, J. R.; Natarajan, V.; Pearse, D. B.; Garcia, J. G.; Verin,A. D. Signaling pathways involved in adenosine triphosphate-inducedendothelial cell barrier enhancement. Circ. Res. 97:115–124; 2005.

[104] Gunduz, D.; Hirche, F.; Hartel, F. V.; Rodewald, C. W.; Schafer, M.;Pfitzer, G.; Piper, H. M.; Noll, T. ATP antagonism of thrombin-inducedendothelial barrier permeability. Cardiovasc. Res. 59:470–478; 2003.

[105] Shinozaki, Y.; Koizumi, S.; Ishida, S.; Sawada, J.; Ohno, Y.; Inoue, K.Cytoprotection against oxidative stress-induced damage of astrocytes byextracellular ATP via P2Y1 receptors. Glia 49:288–300; 2005.

[106] Inoue, K. ATP receptors for the protection of hippocampal functions. Jpn.J. Pharmacol. 78:405–410; 1998.

[107] Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J. V.; Zuo, Y.; Jung, S.;Littman, D. R.; Dustin, M. L.; Gan, W. B. ATP mediates rapidmicroglial response to local brain injury in vivo. Nat. Neurosci. 8:752–758; 2005.

[108] Yitzhaki, S.; Shneyvays, V.; Jacobson, K. A.; Shainberg, A. Involvement

39S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

of uracil nucleotides in protection of cardiomyocytes from hypoxic stress.Biochem. Pharmacol. 69:1215–1223; 2005.

[109] Ninomiya, H.; Otani, H.; Lu, K.; Uchiyama, T.; Kido, M.; Imamura, H.Complementary role of extracellular ATP and adenosine in ischemicpreconditioning in the rat heart. Am. J. Physiol, Heart Circ. Physiol. 282:H1810–H1820; 2002.

[110] Proctor, R. A.; Denlinger, L. C.; Leventhal, P. S.; Daugherty, S. K.;van de Loo, J. W.; Tanke, T.; Firestein, G. S.; Bertics, P. J. Protectionof mice from endotoxic death by 2-methylthio-ATP. Proc. Natl. Acad.Sci. USA 91:6017–6020; 1994.

[111] Gelain, D. P.; de Souza, L. F.; Bernard, E. A. Extracellular purines fromcells of seminiferous tubules. Mol. Cell. Biochem. 245:1–9; 2003.

[112] Pines, A.; Perrone, L.; Bivi, N.; Romanello, M.; Damante, G.; Gulisano,M.; Kelley, M. R.; Quadrifoglio, F.; Tell, G. Activation of APE1/Ref-1 isdependent on reactive oxygen species generated after purinergic receptorstimulation by ATP. Nucleic Acids Res. 33:4379–4394; 2005.

[113] Lee, Y. J.; Han, H. J. Effect of adenosine triphosphate in renal ischemicinjury: involvement of NF-kappaB. J. Cell. Physiol. 204:792–799;2005.

[114] Varani, J.; Phan, S. H.; Gibbs, D. F.; Ryan, U. S.; Ward, P. A. H2O2-mediated cytotoxicity of rat pulmonary endothelial cells. Changes inadenosine triphosphate and purine products and effects of protectiveinterventions. Lab. Invest. 63:683–689; 1990.

[115] Fredholm, B. B.; Arslan, G.; Halldner, L.; Kull, B.; Schulte, G.;Wasserman, W. Structure and function of adenosine receptors and theirgenes. Naunyn-Schmiedeberg's Arch. Pharmacol. 362:364–374; 2000.

[116] Ohta, A.; Sitkovsky, M. Role of G-protein-coupled adenosine receptors indownregulation of inflammation and protection from tissue damage.Nature 414:916–920; 2001.

[117] Lee, H. T.; Emala, C. W. Preconditioning and adenosine protect humanproximal tubule cells in an in vitro model of ischemic injury. J. Am. Soc.Nephrol. 13:2753–2761; 2002.

[118] Kellerman, D.; Evans, R.; Mathews, D.; Shaffer, C. Inhaled P2Y2receptor agonists as a treatment for patients with cystic fibrosis lungdisease. Adv. Drug Deliv. Rev. 54:1463–1474; 2002.

[119] Bannwarth, B.; Allaert, F. A.; Avouac, B.; Rossignol, M.; Rozenberg, S.;Valat, J. P. A randomized, double-blind, placebo controlled triphosphatein study of oral adenosine subacute low back pain. J. Rheumatol.32:1114–1117; 2005.

[120] Hayashida, M.; Fukuda, K.; Fukunaga, A.; Meno, A.; Sato, K.; Tarui, K.;Arita, H.; Kaneko, Y.; Hanaoka, K. Analgesic effect of intravenous ATPon postherpetic neuralgia in comparison with responses to intravenousketamine and lidocaine. J. Anesth. 19:31–35; 2005.

[121] Rapp, D. E.; Lyon, M. B.; Bales, G. T.; Cook, S. P. A role for the P2Xreceptor in urinary tract physiology and in the pathophysiology of urinarydysfunction. Eur. Urol. 48:303–308; 2005.

[122] Hillman, K. A.; Woolf, A. S.; Johnson, T. M.; Wade, A.; Unwin, R. J.;Winyard, P. J. The P2X7 ATP receptor modulates renal cyst developmentin vitro. Biochem. Biophys. Res. Commun. 322:434–439; 2004.

[123] Bean, B. P. Pharmacology and electrophysiology of ATP-activated ionchannels. Trends Pharmacol. Sci. 13:87–90; 1992.

[124] Dubyak, G. R.; el-Moatassim, C. Signal transduction via P2-purinergicreceptors for extracellular ATP and other nucleotides. Am. J. Physiol.265:C577–C606; 1993.

[125] Ralevic, V.; Burnstock, G. Receptors for purines and pyrimidines.Pharmacol. Rev. 50:413–492; 1998.

[126] Communi, D.; Janssens, R.; Suarez-Huerta, N.; Robaye, B.; Boeynaems,J. M. Advances in signalling by extracellular nucleotides. the role andtransduction mechanisms of P2Y receptors. Cell. Signal. 12:351–360;2000.

[127] Dorn, C. C.; Rice, W. R.; Singleton, F. M. Calcium mobilization andresponse recovery following P2-purinoceptor stimulation of rat isolatedalveolar type II cells. Br. J. Pharmacol. 97:163–170; 1989.

[128] Homolya, L.;Watt,W. C.; Lazarowski, E. R.; Koller, B. H.; Boucher, R. C.Nucleotide-regulated calcium signaling in lung fibroblasts and epithelialcells from normal and P2Y(2) receptor (−/−) mice. J. Biol. Chem.274:26454–26460; 1999.

[129] Sienaert, I.; Huyghe, S.; Parys, J. B.; Malfait, M.; Kunzelmann, K.; De

Smedt, H.; Verleden, G. M.; Missiaen, L. ATP-induced Ca2+ signals inbronchial epithelial cells. Pflugers Arch. 436:40–48; 1998.

[130] Seger, R.; Krebs, E. G. The MAPK signaling cascade. FASEB J.9:726–735; 1995.

[131] Toker, A.; Newton, A. C. Cellular signaling: pivoting around PDK-1.Cell103:185–188; 2000.

[132] Raught, B.; Gingras, A. C.; Sonenberg, N. The target of rapamycin (TOR)proteins. Proc. Natl. Acad. Sci. USA 98:7037–7044; 2001.

[133] Wang, X.; Arcuino, G.; Takano, T.; Lin, J.; Peng, W. G.; Wan, P.; Li, P.;Xu, Q.; Liu, Q. S.; Goldman, S. A.; Nedergaard, M. P2X7 receptorinhibition improves recovery after spinal cord injury. Nat. Med.10:821–827; 2004.

[134] Bivalacqua, T. J.; Champion, H. C.; Shah, M. K.; De Witt, B. J.; Inscho,E. W.; Kadowitz, P. J. Comparative responses to alpha,beta-methylene-ATP in cat pulmonary, mesenteric, and hindquarter vascular beds. J. Appl.Physiol. 93:1287–1295; 2002.

[135] Liu, S. F.; McCormack, D. G.; Evans, T. W.; Barnes, P. J. Evidence fortwo P2-purinoceptor subtypes in human small pulmonary arteries. Br. J.Pharmacol. 98:1014–1020; 1989.

[136] Liu, S. F.; McCormack, D. G.; Evans, T. W.; Barnes, P. J.Characterization and distribution of P2-purinoceptor subtypes in ratpulmonary vessels. J. Pharmacol. Exp. Ther. 251:1204–1210; 1989.

[137] Yamamoto, K.; Korenaga, R.; Kamiya, A.; Qi, Z.; Sokabe, M.; Ando, J.P2X(4) receptors mediate ATP-induced calcium influx in human vascularendothelial cells. Am. J. Physiol, Heart Circ. Physiol. 279:H285–H292;2000.

[138] Lewis, C. J.; Evans, R. J. P2X receptor immunoreactivity in differentarteries from the femoral, pulmonary, cerebral, coronary and renalcirculations. J. Vasc. Res. 38:332–340; 2001.

[139] Nori, S.; Fumagalli, L.; Bo, X.; Bogdanov, Y.; Burnstock, G. Coexpres-sion of mRNAs for P2X1, P2X2 and P2X4 receptors in rat vascularsmooth muscle: an in situ hybridization and RT-PCR study. J. Vasc. Res.35:179–185; 1998.

[140] McMillan, M. R.; Burnstock, G.; Haworth, S. G. Vasoconstriction ofintrapulmonary arteries to P2-receptor nucleotides in normal andpulmonary hypertensive newborn piglets. Br. J. Pharmacol. 128:549–555; 1999.

[141] Liang, L.; Zsembery, A.; Schwiebert, E. M. RNA interference targeted tomultiple P2X receptor subtypes attenuates zinc-induced calcium entry.Am. J. Physiol., Cell Physiol. 289:C388–C396; 2005.

[142] Sim, J. A.; Young, M. T.; Sung, H. Y.; North, R. A.; Surprenant, A.Reanalysis of P2X7 receptor expression in rodent brain. J. Neurosci.24:6307–6314; 2004.

[143] Chen, Z.; Jin, N.; Narasaraju, T.; Chen, J.; McFarland, L. R.; Scott,M.; Liu, L. Identification of two novel markers for alveolar epithelialtype I and II cells. Biochem. Biophys. Res. Commun. 319:774–780;2004.

[144] Wolff, S. C.; Qi, A. D.; Harden, T. K.; Nicholas, R. A. Polarizedexpression of human P2Y receptors in epithelial cells from kidney,lung, and colon. Am. J. Physiol., Cell Physiol. 288:C624–C632;2005.

[145] Konduri, G. G.; Bakhutashvili, I.; Frenn, R.; Chandrasekhar, I.; Jacobs,E. R.; Khanna, A. K. P2Y purine receptor responses and expression inthe pulmonary circulation of juvenile rabbits. Am. J. Physiol. HeartCirc. Physiol. 287:H157–H164; 2004.

[146] Communi, D.; Paindavoine, P.; Place, G. A.; Parmentier, M.; Boeynaems,J. M. Expression of P2Y receptors in cell lines derived from the humanlung. Br. J. Pharmacol. 127:562–568; 1999.

[147] Ito, Y.; Son, M.; Sato, S.; Ishikawa, T.; Kondo, M.; Nakayama, S.;Shimokata, K.; Kume, H. ATP release triggered by activation of the Ca2+-activated K+channel in human airway Calu-3 cells. Am. J. Respir. CellMol. Biol. 30:388–395; 2004.

[148] Balestrieri, M. L.; Malik, K. U.; Balestrieri, C.; Lee, T. C. Types ofpurinoceptors and phospholipase A2 involved in the activation of theplatelet-activating factor-dependent transacetylase activity and arachido-nate release by ATP in endothelial cells. Prostaglandins Other LipidMediat. 56:363–375; 1998.

[149] Chen, B. C.; Lee, C. M.; Lee, Y. T.; Lin, W. W. Characterization of

40 S. Ahmad et al. / Free Radical Biology & Medicine 41 (2006) 29–40

signaling pathways of P2Y and P2U purinoceptors in bovine pulmonaryartery endothelial cells. J. Cardiovasc. Pharmacol. 28:192–199; 1996.

[150] Govindaraju, V.; Martin, J. G.; Maghni, K.; Ferraro, P.; Michoud, M. C.The effects of extracellular purines and pyrimidines on human airwaysmooth muscle cells. J. Pharmacol. Exp. Ther. 315:941–948; 2005.

[151] Clunes, M. T.; Collett, A.; Baines, D. L.; Bovell, D. L.; Murphie, H.;Inglis, S. K.; McAlroy, H. L.; Olver, R. E.; Wilson, S. M. Culturesubstrate-specific expression of P2Y2 receptors in distal lung epithelialcells isolated from foetal rats. Br. J. Pharmacol. 124:845–847; 1998.

[152] Gobran, L. I.; Rooney, S. A. Surfactant secretagogue activation of protein

kinase C isoforms in cultured rat type II cells. Am. J. Physiol. 277:L251–L256; 1999.

[153] Bergner, A.; Sanderson, M. J. ATP stimulates Ca2+ oscillations andcontraction in airway smooth muscle cells of mouse lung slices. Am. J.Physiol., Lung Cell Mol. Physiol. 283:L1271–L1279; 2002.

[154] Muller, T.; Bayer, H.; Myrtek, D.; Ferrari, D.; Sorichter, S.;Ziegenhagen, M. W.; Zissel, G.; Virchow Jr., J. C.; Luttmann, W.;Norgauer, J.; Di Virgilio, F.; Idzko, M. The P2Y14 receptor of airwayepithelial cells: coupling to intracellular Ca2+ and IL-8 secretion. Am.J. Respir. Cell Mol. Biol. 33:601–609; 2005.


Recommended