Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 19.
Published in final edited form as: Inflammation. 2012 Apr;35(2):614–622. doi: 10.1007/s10753-011-9353-1

The Effect of A2A Adenosine Receptor Activation on C-C Chemokine Receptor 7 Expression in Human THP1 Macrophages During Inflammation

Adrienne J Williams 1, Bruce N Cronstein 1,2,3
PMCID: PMC4366145  NIHMSID: NIHMS669900  PMID: 21739164

Abstract

C-C chemokine receptor 7 (CCR7) and its chemoattractant agonist CCL21 promote cell migration and expression of pro-inflammatory proteins in an atherogenic environment. Since A2A adenosine receptor activation reduces migration and inflammatory effects, we examined its effect on CCR7 expression and migration. CCR7 protein expression decreased by about a third in macrophages treated with A2A receptor agonist CGS 21680 (p=0.028, n=7) and was reversed with antagonist, although mRNA levels increased twofold (p=0.001, n=3). Furthermore, macrophages treated with CGS 21680 showed a significant decrease in migration (p=0.0311, n=7). These results suggest that A2A adenosine receptor activation not only modulates CCR7 expression in both normal and inflammatory environments but also regulates macrophage migration to CCR7-specific chemoattractants.

Keywords: A2A adenosine receptor, macrophage, chemokine receptor migration

INTRODUCTION

Atherogenesis occurs when arterial injury leads to a persistent inflammatory response—inflammatory cells respond and migrate to the area, macrophages take up oxidized low-density lipoprotein and, in the process, become foam cells, undergo apoptosis, and accumulate around the lesion, forming a growing plaque that undergoes ineffective repair and may calcify, becoming a complex plaque [14]. Chemokines and their receptors play a role in atherogenesis and arterial repair by regulating the recruitment and function of pro-inflammatory and plaque-destabilizing cells [57].

The purine adenosine accumulates during tissue injury or stress and, like chemokines, can modulate immune response and cellular function through its receptor subtype A2A [811]. A2A adenosine receptor activation suppresses inflammation by regulating cytokine production and prevents atherogenesis by suppressing cell migration and reducing foam cell formation through reverse cholesterol transport [1217]. The A2A adenosine receptor also modulates chemokine receptor levels; in particular, it downregulates C-C chemokine receptor subtype 7 (CCR7) expression and function in T and dendritic cells [1820]. The attraction of monocytes and macrophages to CCR7’s agonists could be used to prevent atherogenesis—if fewer new macrophages migrate into the atherosclerotic plaque, further arterial injury may be prevented.

We hypothesized that A2A adenosine receptor activation modulates CCR7 macrophage expression and migration. We found that while A2A adenosine receptor stimulation increased CCR7 mRNA, it decreased protein expression in both noninflammatory and inflammatory environments. As a result, chemotaxis of cells to CCR7 agonists decreased. We also examined signaling at A2A receptors for regulation of CCR7 receptors and found that, as with other functions, adenosine A2A receptors regulate CCR7 expression by a pathway dependent on PKA and MAPK.

MATERIALS AND METHODS

Materials

The selective A2A receptor agonist CGS21680 and its antagonist ZM 241385 were purchased from Tocris Cookson (Bristol, UK). The selective CCR7 agonists CCL19 and CCL21 and the cytokine IFNγ were obtained from R&D Systems (Minneapolis, MN). Epac agonist 8-pcPT-2′-O-Me-cAMP was purchased from Axxora Diagnostics (San Diego, CA) and PKA inhibitor PKI, corresponding to amino acids 5 through 24, from Promega (Madison, WI). Rac inhibitor 553502 and p38 inhibitor SB202190 were obtained from Calbiochem (San Diego, CA), and Mek inhibitor U0126 from Sigma-Aldrich (St. Louis, MO).

Cell Culture

Human monocytic THP1 cells were purchased from American Type Culture Collection (Manassas, VA). Cells were grown in RPMI-1640 media supplemented with 10% FBS, 1% penicillin and streptomycin, and 1% l-glutamine (Invitrogen, Carlsbad, CA) at 37°C, 5% CO2. Cells were differentiated to macrophages by incubation with 25 ng/mL phorbol 12-myristate 13-acetate (Sigma-Aldrich) for 48 h.

RNA Isolation and Real-Time RT-PCR

THP1 monocytes were differentiated to macrophages (700,000 cells per milliliter), given fresh media, and treated with combinations of CGS21680 (1 µM and 100 nM) and ZM241385 (10 µM) for 4 h. For inflammatory experiments, cells were pretreated with 500 U/mL IFNγ for 12 to 16 h. For signaling experiments, cells were pretreated for 1 h with PKA inhibitor (10 µM), p38 inhibitor (10 µM), Rac inhibitor (10 µM), or Mek inhibitor (10 µM). Media were then removed, and RNA was extracted using Trizol (Invitrogen) according to the manufacturer’s instructions. Of total RNA, 1.5 µg was used from each sample for first-strand, complementary deoxyribonucleic acid synthesis with random hexamer primers using an RNA PCR Core Kit (Applied Biosystems, Foster City, CA).

For quantification by real-time PCR, the following primer pairs were used: CCR7 F: 5′-TGACATGCACTCAGCTCTTG-3 R: 5′-AGGTTTTCAGTCCCTGTGAC-3′ (60°C, 136 bp). GAPDH F: 5′-AACATCATCCCTGCCTCTAC-3′ R: 5′-CCCTCTTGCTGATGCCAAAT-3′ (58°C, 358 bp). PCR product was detected using SYBR Green and the MxPro 3005P from Agilent Technologies (Cedar Creek, TX). Gene expression was quantified to a standard curve and normalized to GAPDH.

Western Blot Analysis

Monocytes were differentiated to macrophages (1.5 million cells per milliliter), given fresh media, and treated with the appropriate cytokines, agonists, and inhibitors for 48 h. Cells were then lysed using radio-immunoprecipitation assay buffer with protease inhibitor cocktail (Sigma-Aldrich) for 30 min at 4°C, centrifuged at 10,000 rpm for 10 min, and the supernatant recovered. Of protein, 35 µg was separated by 10% SDS–PAGE and electrotransferred to nitrocellulose membranes (BioRad, Hercules, CA). Nonspecific antibody binding was blocked with Superblock T20 (Thermo, Rockford, IL) for 4 h at room temperature. The membrane was incubated with a primary antibody against CCR7 (goat polyclonal from Santa Cruz Biotechnologies, Santa Cruz, CA) overnight at 4°C and then incubated with anti-goat secondary antibody conjugated to alkaline phosphatase (Santa Cruz Biotechnologies) for 1 h at room temperature, after washing with TTBS. Protein bands were visualized using electrochemifluorescence substrate and a Storm Phosphoimager (GE Healthcare). Then, the membrane was stripped and incubated with a primary antibody against beta-actin (mouse monoclonal from Abcam, Cambridge, MA) to be used as an internal, normalizing standard. Band intensity was quantified using Kodak Molecular Imaging software (Eastman Kodak, Rochester, NY).

In Vitro Chemotaxis Cell Migration Assay

Monocytes were differentiated into macrophages, given fresh media, and pretreated with the appropriate cytokines, agonists, and antagonists for 48 h. Two hundred thousand cells were then gently lifted using rubber scrapers and Dispase (Roche, Mannheim, Germany) and added to 5-µm chamber inserts cells from the QCM Cell Migration Assay Kit (Chemicon, Temecula, CA) with fresh media containing 1% FBS, 1% penicillin–streptomycin, and 1% l-glutamine in RPMI-1640 media (Invitrogen); 300 ng/mL CCL19 or CCL21 was added to starved media in the lower chamber as a chemoattractant. Cells were then incubated for 24 h, and contents of the lower chamber were detached, stained with fluorescein, and lysed according to the manufacturer’s instructions. Migration was quantified using the Victor3V Plate Reader (Perkin Elmer).

Statistics

Data are reported as the mean±SEM. Differences between treatment groups were analyzed with Graph-Pad Prism (GraphPad Software, San Diego, CA) using one-way analysis of variance followed by Tukey posttest, paired and unpaired Student’s t tests. A p value of less than 0.05 was considered statistically significant.

RESULTS

A2A Adenosine Receptor Agonist CGS21680 Increases CCR7 mRNA Expression

There was a dose-dependent increase in CCR7 message in macrophages following treatment with the adenosine A2A agonist CGS21680 (p<0.001 vs. control, n=3), and co-treatment with the A2A antagonist ZM241385 abrogated the increase p<0.01 vs. CGS, n=3, Fig. 1a).

Fig. 1.

Fig. 1

A2A adenosine receptor agonist CGS21680 increases CCR7 mRNA expression and is reversed with antagonist ZM241385 in THP1 macrophages. Messenger RNA isolated from cells pretreated overnight in a regular media conditions or b IFNγ (500 U/mL) and then treated with combinations of CGS21680 (10−6 M and 10−7 M), ZM241385 (10−5 M), and DMSO as a negative control for 4 h and were subjected to real-time RT-PCR using specific primers. The expression level was quantified using real-time PCR and normalized to GAPDH. Data shown are the means±SEM of the percentages of control from three independent experiments. a vs. Control, * p=0.001; vs. CGS, #p=0.01; b vs. CGS, #p=0.0382.

Cytokines, which are abundant in atherosclerotic lesions, may affect expression of such genes as A2A adenosine receptor expression in THP1 cells. Indeed, IFNγ diminishes both expression and function of A2A receptors and plays a central role in atherogenesis [13]. We therefore determined whether IFNγ altered A2A-mediated increases in CCR7 mRNA. We were surprised to observe that, unlike other adenosine receptor functions, IFNγ did not alter adenosine A2A-mediated increase in CCR7 message or the capacity of ZM241385 to reverse that effect (p<0.04 vs. CGS, n=3, Fig. 1b).

A2A Adenosine Receptor Agonist CGS21680 Decreases CCR7 Protein Expression

Because A2A receptor stimulation increased mRNA for CCR7, we next determined whether A2A adenosine receptor activation similarly regulated CCR7 protein expression. We were surprised to find that, in contrast to mRNA expression, CCR7 protein levels decreased by about a third (p<0.03 vs. control, n=7), an effect that was reversed by the antagonist (p<0.05 vs. CGS, n=7, Fig. 2a). Pretreatment with the cytokine IFNγ also decreased CCR7 protein levels with CGS21680 treatment (p<0.04 vs. control, n=3) and reversed with ZM241385 (p<0.03 vs. CGS, n=3, Fig. 2b).

Fig. 2.

Fig. 2

A2A adenosine receptor agonist CGS21680 decreases CCR7 protein expression and is reversed with antagonist ZM241385 in THP1 macrophages. Cells pretreated overnight in a regular media conditions or b IFNγ (500 U/mL) and then treated with combinations of CGS21680 (10−6 M and 10−7 M), ZM241385 (10−5 M), and DMSO as a negative control for 48 h had 35 µg of their protein lysate analyzed with SDS–PAGE and immunoblotting. c Representative immunoblots. The expression level was quantified using Kodak Image Analysis software and normalized to beta-actin. Data shown are the means±SEM of the percentages of control from a seven blots and b three blots. a vs. Control, *p=0.028; vs. CGS, #p=0.0432; b vs. Control, *p=0.0386, **p=0.0085; vs. CGS, #p=0.0234.

CGS21680 Modulates CCR7-Mediated Migration in Normal and Pro-inflammatory Environments

Prior work has indicated that adenosine A2A receptor stimulation retards chemotactic migration to CCR7 ligands by dendritic cells, and our studies indicate that adenosine A2A receptor stimulation diminishes CCR7 expression on THP-1 cells [18]. We therefore determined whether adenosine A2A receptor stimulation regulates chemotaxis to CCR7 ligands by THP-1 cells in the absence or presence of inflammation. CGS21680 pretreatment reduced migration by 17% to CCL21 (p<0.03 vs. control, n=7) and by a similar amount after IFNγ pretreatment (p<0.03 vs. IFNγ, n=7, Fig. 3). Migration to CCL19, a ligand for CCR7, was also reduced. As with the effects on CCR7 expression, the effects of the adenosine receptor agonist were reversed by the A2A receptor antagonist ZM241385 (p<0.008 vs. CGS, n=7, Fig. 4).

Fig. 3.

Fig. 3

CGS21680 treatment with pro-inflammatory cytokines decreases THP1 macrophage migration to CCR7 agonists. Cells were pretreated overnight with IFNγ (500 U/mL) or TNFα (10 ng/mL) and then with CGS21680 (10−6 M) or DMSO as a negative control for 48 h. Cells were then lifted, added in suspension to 5-µm upper chamber inserts, and migrated to a lower chamber with a CCL19 (300 ng/mL) or b CCL21 (300 ng/mL) for 24 h. Migration was quantified using fluorescein labeling of cells. Data shown are the means±SEM of the percentages of three independent experiments with CCL19 and six with CCL21. vs. Control, * p=0.0311, ** p=0.0389, *** p=0.0196. vs. IFNγ, #p=0.0273.

Fig. 4.

Fig. 4

CGS21680 treatment with and without IFNγ decreases THP1 macrophage migration to CCR7 agonists and is reversed with antagonist ZM241385. Cells were first pretreated overnight with IFNγ (500 U/mL) or TNFα (10 ng/mL) and then with combinations of CGS21680 (10−6 M), ZM241385 (10−5 M), or DMSO as a negative control for 48 h. Cells were then lifted, added in suspension to 5-µm upper chamber inserts, and migrated to a lower chamber with CCL21 (300 ng/mL) for 24 h. Migration was quantified using fluorescein labeling of cells. Data shown are the means±SEM of the percentages of six independent experiments—three with ZM. vs. Control, *p=0.0311, **p=0.0273. vs. CGS, #p=0.0078, ##p=0.125.

A2A Adenosine Receptor-Induced Changes in CCR7 Expression Are Blocked with PKA, p38, and Mek Inhibitors

A2A adenosine receptors stimulate adenylate cyclase activity and increase cellular cAMP content, which activates Epac and/or PKA, which activate signaling pathways including MAPK and p38 [21, 22]. To probe the signaling pathways involved, we used pharmacologic inhibitors of protein kinase A and erk1/2 and p38 MAP kinases and observed their effect on the capacity of adenosine A2A receptor stimulation to alter mRNA levels for CCR7. We found inhibitors of PKA and Mek, but not a p38MAPK inhibitor or an inhibitor of rac (Fig. 5a, b). In contrast, p38 and rac inhibition completely blocked the effect of A2A receptor stimulation on CCR7 protein expression (p<0.05 vs. CGS with p38 inhibition, n=4, Fig. 5c, d).

Fig. 5.

Fig. 5

A2A adenosine receptor-induced increase of CCR7 mRNA expression and decrease of CCR7 protein expression in THP1 macrophages can be reversed with PKA, p38, and Mek inhibitors. Cells were treated with combinations of a 10−6 M or b 10−7 M CGS21680 with p38i (10 µM), PKAi (10 µM), Meki (10 µM), Raci (10 µM), Epac analog (10 µM), and DMSO as a negative control for 4 h, and the messenger RNA collected and subjected to real-time RT-PCR using specific primers. The expression level was quantified using real-time PCR and normalized to GAPDH. Data shown are the means±SEM of the percentages of control from three independent experiments. Cells were also treated with combinations of c 10−6 M or d 10−7 M CGS21680 and inhibitors for 48 h and had 35 µg of their cell lysate analyzed with SDS–PAGE and immunoblotting. e Representative immunoblots. The expression level was quantified using Kodak Image Analysis software and normalized to beta-actin. Data shown are the means±SEM of the percentages of control from three to four blots. a vs. Control, *p=0.065, **p=0.0416; b vs. Control, *p=0.043; c vs. Control, *p=0.038; vs. CGS, #p=0.0504; d vs. Control, *p=0.0581; vs. CGS, #p=0.0204.

DISCUSSION

Since A2A adenosine receptor stimulation reduces inflammatory and atherogenic effects and CCR7 activation promotes these functions, we hypothesized that A2A adenosine receptor activation could reduce CCR7 expression and thus modulate macrophage migration to inflamed sites or atherosclerotic plaques. Our results provide very interesting results regarding the effects of A2A receptor ligation on CCR7 expression. We found that receptor ligation increases CCR7 mRNA by a PKA- and MEK-dependent pathway but diminishes surface expression of the protein by a p38MAPK-, rac-dependent pathway (Fig. 6). Clearly, the resulting reduction in CCR7 chemotactic function results from the diminished receptor expression/function.

Fig. 6.

Fig. 6

The A2A adenosine receptor uses signaling pathways downstream of PKA to regulate CCR7 expression. The A2A adenosine receptor subtype’s G-stimulatory protein subunit (Gs) activates adenylate cyclase (AC), which leads to production of cAMP and stimulation of primarily p38 signaling to decrease CCR7 protein and primarily MA-PK signaling to increase CCR7 mRNA in THP1 macrophages.

Macrophages near or within an atherosclerotic plaque are exposed to inflammatory cytokines like IFNγ, which diminish A2A adenosine receptor expression and function and might influence their ability to regulate CCR7 levels [13, 23]. Furthermore, reverse cholesterol transport experiments demonstrate that A2A adenosine receptor function in macrophages is altered by IFNγ [12, 15, 17, 24]. Unlike other adenosine A2A receptor-mediated effects, the A2A receptor-stimulated effects on CCR7 mRNA and protein expression were unaffected by IFNγ.

The contrast between adenosine receptor-stimulated increases in CCR7 mRNA and decreases in CCR7 protein expression and function were surprising. One explanation for these observations is that chemokine receptor processing and transport via the endoplasmic reticulum and Golgi apparatus may be disrupted, degraded, or downgraded by adenosine A2A receptor stimulation. Adenosine A2A receptor stimulation may also increase the rate at which CCR7 receptors are cleared from the surface of the cell, as previously demonstrated for other chemotactic receptors in neutrophils [2527]. Alternatively, translation of CCR7 message may be blocked by adenosine receptor-stimulated microRNAs; a database search has found some miRNAs with sequence similarity of more than 65% with CCR7, although they are not located in the 5′ region considered most effective for translational blocking (mirbase.org) [2831].

To determine a signal transduction pathway between A2A adenosine receptor activation and CCR7 expression, we tested pharmacologic signal transduction inhibitors corresponding to previously demonstrated adenosine A2A receptor signaling pathways. A2A adenosine receptor stimulation can activate either the signaling proteins Epac or PKA [17, 32, 33]. PKA activation may then influence many possible downstream pathways, most commonly Erk with its upstream protein Mek and p38 with its upstream protein Rac [21, 34, 35]. We found PKA inhibition reversed A2A adenosine receptor-induced changes in CCR7 message, while Epac analog treatment failed to mimic expression, indicating a PKA-dependent pathway. Mek inhibition blocked change in CCR7 expression as well, while p38 and Rac inhibition did not affect CCR7 message but did change CCR7 protein expression. These results suggest A2A adenosine receptor activation in THP1 macrophages leads to MAPK signaling through a PKA-dependent pathway for CCR7 expression.

Although we showed the presence of IFNγ did not hinder A2A adenosine receptor’s ability to reduce CCR7 expression and cell migration, the cytokines TNFα and interleukin-1 also affect A2A adenosine receptor expression but were not examined [13]. A thorough examination of the other two cytokines with CGS21680, or a possible combination of all three, would give a better understanding of the receptor’s function in an inflammatory, atherogenic environment.

An increasing body of literature demonstrates that adenosine and its receptors may play a role in the development and regression of atherosclerosis. Prior work demonstrates that adenosine A2A receptor activation diminishes foam cell formation not only by increasing ABCA1 expression but by increasing its function as well [12, 3639]. We now provide further evidence that adenosine and its receptors might play a beneficial role in atherosclerosis by inhibiting recruitment of macrophages to atherosclerotic plaques.

ABBREVIATIONS

cAMP

Cyclic adenosine monophosphate

CCL19

C-C chemokine ligand 19

CCL21

C-C chemokine ligand 21

CCR7

C-C chemokine receptor subtype 7

CGS21680

2-p-(2-carboxyethyl) phenethyllamino-5’-N-ethylcarboxamido- adenosine

DMSO

Dimethyl sulfoxide

Epac

Guanine nucleotide exchange factor activated by cAMP

Erk

Extracellular signal regulated kinase

FBS

Fetal bovine serum

GAPDH

Glyceraldehyde phosphate dehydrogenase

IFNγ

Interferon gamma

MAPK

Mitogen-activated protein kinase

Mek

MAP kinase kinase

p38

p38 mitogen-activated protein kinase

PCR

Polymerase chain reaction

PKA

Protein kinase A

Rac

Subfamily of Rho GTPases

RNA

Ribonucleic acid

RPMI-1640

Roswell Park Memorial Institute media

SDS–PAGE

Sodium dodecyl sulfate–polyacrylamide gel electrophoresis

SEM

Standard error mean

SYBR Green

N',N'-dimethyl-N-[4-[(E)-(3-methyl-1,3-benzothiazol-2-ylidene)methyl]-1-phenylquinolin-1-ium-2-yl]-N-propylpropane-1,3-diamine

THP1

Human acute monocytic leukemia cell line

TNFα

Tumor necrosis factor alpha

TTBS

Tween/Tris-buffered saline ZM 241385–4-(2-[7-amino- 2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol

REFERENCES

  • 1.Lord RS, Bobryshev YV. Hallmarks of atherosclerotic lesion development with special reference to immune inflammatory mechanisms. Cardiovascular Surgery. 2002;10:405–414. doi: 10.1016/s0967-2109(02)00039-x. [DOI] [PubMed] [Google Scholar]
  • 2.Lusis AJ. Atherosclerosis. Nature. 2000;407:233–241. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Steinberg D. Atherogenesis in perspective: hypercholesterolemia and inflammation as partners in crime. Nature Medicine. 2002;8:1211–1217. doi: 10.1038/nm1102-1211. [DOI] [PubMed] [Google Scholar]
  • 4.Takahashi K, Takeya M, Sakashita N. Multifunctional roles of macrophages in the development and progression of atherosclerosis in humans and experimental animals. Medical Electron Microscopy. 2002;35:179–203. doi: 10.1007/s007950200023. [DOI] [PubMed] [Google Scholar]
  • 5.Campbell DJ, Kim CH, Butcher EC. Chemokines in the systemic organization of immunity. Immunological Reviews. 2003;195:58–71. doi: 10.1034/j.1600-065x.2003.00067.x. [DOI] [PubMed] [Google Scholar]
  • 6.Lucas AD, Greaves DR. Atherosclerosis: role of chemokines and macrophages. Expert Reviews in Molecular Medicine. 2001;3:1–18. doi: 10.1017/S1462399401003696. [DOI] [PubMed] [Google Scholar]
  • 7.Zlotnik A, Yoshie O, Nomiyama H. The chemokine and chemokine receptor superfamilies and their molecular evolution. Genome Biology. 2006;7:243. doi: 10.1186/gb-2006-7-12-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN, Dagnelie PC. Adenosine 5'-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation. Pharmacology & Therapeutics. 2006;112:358–404. doi: 10.1016/j.pharmthera.2005.04.013. [DOI] [PubMed] [Google Scholar]
  • 9.Hasko G, Pacher P, Deitch EA, Vizi ES. Shaping of monocyte and macrophage function by adenosine receptors. Pharmacology & Therapeutics. 2007;113:264–275. doi: 10.1016/j.pharmthera.2006.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.McPherson JA, Barringhaus KG, Bishop GG, Sanders JM, Rieger JM, Hesselbacher SE, Gimple LW, Powers ER, Macdonald T, Sullivan G, et al. Adenosine A(2A) receptor stimulation reduces inflammation and neointimal growth in a murine carotid ligation model. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21:791–796. doi: 10.1161/01.atv.21.5.791. [DOI] [PubMed] [Google Scholar]
  • 11.Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death and Differentiation. 2007;14:1315–1323. doi: 10.1038/sj.cdd.4402132. [DOI] [PubMed] [Google Scholar]
  • 12.Bingham TC, Fisher EA, Parathath S, Reiss AB, Chan ES, Cronstein BN. A2A adenosine receptor stimulation decreases foam cell formation by enhancing ABCA1-dependent cholesterol efflux. Journal of Leukocyte Biology. 2010;87:683–690. doi: 10.1189/jlb.0709513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Khoa ND, Montesinos MC, Reiss AB, Delano D, Awadallah N, Cronstein BN. Inflammatory cytokines regulate function and expression of adenosine A(2A) receptors in human monocytic THP-1 cells. Journal of Immunology. 2001;167:4026–4032. doi: 10.4049/jimmunol.167.7.4026. [DOI] [PubMed] [Google Scholar]
  • 14.Link AA, Kino T, Worth JA, McGuire JL, Crane ML, Chrousos GP, Wilder RL, Elenkov IJ. Ligand-activation of the adenosine A2a receptors inhibits IL-12 production by human monocytes. Journal of Immunology. 2000;164:436–442. doi: 10.4049/jimmunol.164.1.436. [DOI] [PubMed] [Google Scholar]
  • 15.Reiss AB, Awadallah NW, Malhotra S, Montesinos MC, Chan ES, Javitt NB, Cronstein BN. Immune complexes and IFN-gamma decrease cholesterol 27-hydroxylase in human arterial endothelium and macrophages. Journal of Lipid Research. 2001;42:1913–1922. [PubMed] [Google Scholar]
  • 16.Thiel M, Chouker A. Acting via A2 receptors, adenosine inhibits the production of tumor necrosis factor-alpha of endotoxin-stimulated human polymorphonuclear leukocytes. The Journal of Laboratory and Clinical Medicine. 1995;126:275–282. [PubMed] [Google Scholar]
  • 17.Block ET. Interferon gamma inhibits adenosine A2A receptor function in hepatic stellate cells by downregulating expression of adenylyl cyclase in a STAT1-dependent manner. New York: New York University Department of Basic Medical Sciences; 2009. [Google Scholar]
  • 18.Hofer S, Ivarsson L, Stoitzner P, Auffinger M, Rainer C, Romani N, Heufler C. Adenosine slows migration of dendritic cells but does not affect other aspects of dendritic cell maturation. The Journal of Investigative Dermatology. 2003;121:300–307. doi: 10.1046/j.1523-1747.2003.12369.x. [DOI] [PubMed] [Google Scholar]
  • 19.Sun J, Zhang Y, Yang M, Zhang Y, Xie Q, Li Z, Dong Z, Yang Y, Deng B, Feng A, et al. Hypoxia induces T-cell apoptosis by inhibiting chemokine C receptor 7 expression: the role of adenosine receptor A(2) Cellular & Molecular Immunology. 2010;7:77–82. doi: 10.1038/cmi.2009.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhang H, Park Y, Wu J, Chen X, Lee S, Yang J, Dellsperger KC, Zhang C. Role of TNF-alpha in vascular dysfunction. Clinical Science (London) 2009;116:219–230. doi: 10.1042/CS20080196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fredholm BB, Chern Y, Franco R, Sitkovsky M. Aspects of the general biology of adenosine A2A signaling. Progress in Neurobiology. 2007;83:263–276. doi: 10.1016/j.pneurobio.2007.07.005. [DOI] [PubMed] [Google Scholar]
  • 22.Schulte G, Fredholm BB. Signalling from adenosine receptors to mitogen-activated protein kinases. Cellular Signalling. 2003;15:813–827. doi: 10.1016/s0898-6568(03)00058-5. [DOI] [PubMed] [Google Scholar]
  • 23.Nguyen DK, Montesinos MC, Williams AJ, Kelly M, Cronstein BN. Th1 cytokines regulate adenosine receptors and their downstream signaling elements in human microvascular endothelial cells. Journal of Immunology. 2003;171:3991–3998. doi: 10.4049/jimmunol.171.8.3991. [DOI] [PubMed] [Google Scholar]
  • 24.Reiss AB, Carsons SE, Anwar K, Rao S, Edelman SD, Zhang H, Fernandez P, Cronstein BN, Chan ES. Atheroprotective effects of methotrexate on reverse cholesterol transport proteins and foam cell transformation in human THP-1 monocyte/macrophages. Arthritis and Rheumatism. 2008;58:3675–3683. doi: 10.1002/art.24040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cronstein BN, Haines KA, Kolasinski S, Reibman J. Gs linked receptors (Beta-adrenergic and adenosine A2) uncouple chemoattractant receptors from G proteins. Clinical Research. 1991;39:343A. [Google Scholar]
  • 26.Cronstein BN, Haines KA, Kolasinski S, Reibman J. Occupancy of G alpha s-linked receptors uncouples chemo-attractant receptors from their stimulus-transduction mechanisms in the neutrophil. Blood. 1992;80:1052–1057. [PubMed] [Google Scholar]
  • 27.Cronstein BN, Rose FR, Pugliese C. Adenosine, a cytoprotective autocoid: effects of adenosine on neutrophil plasma membrane viscosity and chemoattractant receptor display. Biochimica et Biophysica Acta. 1989;987:176–180. doi: 10.1016/0005-2736(89)90541-5. [DOI] [PubMed] [Google Scholar]
  • 28.The microRNA registry. [Accessed 29 June 2010]; [ mirbase.org]. [Google Scholar]
  • 29.Hawkins PG, Morris KV. RNA and transcriptional modulation of gene expression. Cell Cycle. 2008;7:602–607. doi: 10.4161/cc.7.5.5522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Maziere P, Enright AJ. Prediction of microRNA targets. Drug Discovery Today. 2007;12:452–458. doi: 10.1016/j.drudis.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 31.Williams AE. Functional aspects of animal microRNAs. Cellular and Molecular Life Sciences. 2008;65:545–562. doi: 10.1007/s00018-007-7355-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bos JL. Epac proteins: multi-purpose cAMP targets. Trends in Biochemical Sciences. 2006;31:680–686. doi: 10.1016/j.tibs.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 33.Wolff J, Londos C, Cooper DM. Adenosine receptors and the regulation of adenylate cyclase. Advances in Cyclic Nucleotide Research. 1981;14:199–214. [PubMed] [Google Scholar]
  • 34.Che J, Chan ES, Cronstein BN. Adenosine A2A receptor occupancy stimulates collagen expression by hepatic stellate cells via pathways involving protein kinase A, Src, and extracellular signal-regulated kinases 1/2 signaling cascade or p38 mitogen-activated protein kinase signaling pathway. Molecular Pharmacology. 2007;72:1626–1636. doi: 10.1124/mol.107.038760. [DOI] [PubMed] [Google Scholar]
  • 35.Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nature Reviews. 2008;7:759–770. doi: 10.1038/nrd2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Reiss AB, Anwar K, Merrill JT, Chan ES, Awadallah NW, Cronstein BN, Michael Belmont H, Belilos E, Rosenblum G, Belostocki K, et al. Plasma from systemic lupus patients compromises cholesterol homeostasis: a potential mechanism linking autoimmunity to atherosclerotic cardiovascular disease. Rheumatology International. 2010;30:591–598. doi: 10.1007/s00296-009-1020-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Reiss AB, Patel CA, Rahman MM, Chan ES, Hasneen K, Montesinos MC, Trachman JD, Cronstein BN. Interferon-gamma impedes reverse cholesterol transport and promotes foam cell transformation in THP-1 human monocytes/macrophages. Medical Science Monitor. 2004;10:BR420–BR425. [PubMed] [Google Scholar]
  • 38.Reiss AB, Rahman MM, Chan ES, Montesinos MC, Awadallah NW, Cronstein BN. Adenosine A2A receptor occupancy stimulates expression of proteins involved in reverse cholesterol transport and inhibits foam cell formation in macrophages. Journal of Leukocyte Biology. 2004;76:727–734. doi: 10.1189/jlb.0204107. [DOI] [PubMed] [Google Scholar]
  • 39.Reiss AB, Wan DW, Anwar K, Merrill JT, Wirkowski PA, Shah N, Cronstein BN, Chan ES, Carsons SE. CD36 scavenger receptor expression in THP-1 human monocytes in the presence of lupus plasma: linking autoimmunity and atherosclerosis. Experimental Biology and Medicine (Maywood) 2009;234:354–360. doi: 10.3181/0806-BC-194. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES