Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Dec 1.
Published in final edited form as: Arthritis Rheum. 2008 Dec;58(12):3675–3683. doi: 10.1002/art.24040

Atheroprotective Effects of Methotrexate on Reverse Cholesterol Transport Proteins and Foam Cell Transformation in THP-1 Human Monocytes/Macrophages

Allison B Reiss 1, Steven E Carsons 1, Kamran Anwar 1, Soumya Rao 1, Sari D Edelman 1, Hongwei Zhang 1, Patricia Fernandez 2, Bruce N Cronstein 2, Edwin SL Chan MD 2
PMCID: PMC2599810  NIHMSID: NIHMS68859  PMID: 19035488

Abstract

OBJECTIVE:

To determine whether MTX can overcome the atherogenic effect of COX-2 inhibitors and IFN-γ, both of which suppress cholesterol efflux protein levels and promote foam cell transformation in THP-1 human monocytes/macrophages.

METHODS:

Message and protein level of the reverse cholesterol transport (RCT) proteins cholesterol 27-hydroxylase and ABCA1 in THP-1 cells were evaluated by real-time polymerase chain reaction and immunoblot, respectively. Expression was evaluated in cells incubated in the presence or absence of the COX-2 inhibitor NS398 or IFN-γ with/without MTX. Foam cell transformation of lipid-loaded THP-1 macrophages was detected with oil red O staining and light microscopy.

RESULTS:

MTX increased 27-hydroxylase message and completely blocked NS398-induced downregulation of 27-hydroxylase (112.8±13.1% for NS398+MTX versus 71.1±4.3% for NS398 alone, with untreated as 100%, n=3, p<0.01). MTX also negated COX-2 inhibitor-mediated downregulation of ABCA1. Reversal of inhibitory effects on 27-hydroxylase and ABCA1 in the presence of MTX were blocked by the adenosine A2A receptor-specific antagonist ZM-241385. MTX also prevented NS398 and IFN-γ from increasing transformation of lipid-loaded THP-1 macrophages into foam cells.

CONCLUSIONS:

This study provides evidence supporting the atheroprotective effect of MTX. Through adenosine A2A receptor activation, MTX promotes RCT and limits foam cell formation in THP-1 macrophages. This is the first evidence that any commonly used medication can increase expression of anti-atherogenic RCT proteins and can counteract the effects of COX-2 inhibition. Our results suggest that one mechanism by which MTX protects against cardiovascular mortality in RA patients is through facilitation of cholesterol outflow from cells of the artery wall.

Introduction

Methotrexate (MTX) has a long history of use in the treatment of various immunologic diseases, and has been used to treat rheumatoid arthritis and psoriasis since the 1960s (1-3). MTX was originally developed as a folate antagonist that inhibits dihydrofolate reductase activity and is used in high doses for the treatment of malignancies such as leukemia. Because of its anti-inflammatory and immunosuppressive effects, the drug is also efficacious in the treatment of diseases such as asthma, systemic lupus erythematosus, Crohn's disease, myositis, and vasculitis (4-8).

A number of clinical studies show that MTX not only inhibits inflammatory cell proliferation through action on dihydrofolate reductase but also inhibits the conversion of 5-aminoimidazole-4-carboxamide ribonucleotide to 5-formyl-5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), thus increasing intracellular and extracellular levels of adenosine-5′-phosphate and adenosine (9, 10). Adenosine is a nucleoside that acts as a signaling molecule by triggering activation of adenosine receptors. These receptors are expressed on the surface of a wide variety of cells and are implicated in cellular protection against ischemia-reperfusion injury (11) and anoxia (12).

The risk of cardiovascular disease (CVD) is increased in patients with rheumatoid arthritis (RA) (13-15) and this elevated risk is not explained by traditional risk factors alone (16). Suggested explanations involve the inflammatory response that characterizes active RA and adverse effects of glucocorticoid therapy or other medications. Previous studies suggest that MTX has a beneficial effect on cardiovascular mortality that is not observed with other antirheumatic drugs (17, 18).

We previously reported that immune reactants interfere with cellular defense against cholesterol overload by diminishing expression of two proteins responsible for reverse transport of cholesterol out of the cell to the circulation for ultimate excretion: cholesterol 27-hydroxylase and ATP binding cassette transporter A1 (ABCA1) (19, 20). The atherosclerosis-promoting cytokine IFN-γ decreased 27-hydroxylase and ABCA1 message and protein expression in THP-1 human monocytes/macrophages. Further, IFN-γ-treated THP-1 macrophages exposed to acLDL formed foam cells more rapidly and in greater proportion than untreated control cells. Most recently, we have shown that inhibition of cyclooxygenase (COX) in THP-1 monocytes/macrophages acts in a pro-atherogenic manner by dose-dependently decreasing 27-hydroxylase and ABCA1 (21). THP-1 macrophages show a significant increase in foam cell transformation in the presence of the COX-2 selective inhibitor NS398 compared to control (21). This work suggests that compromise of reverse cholesterol transport may contribute to the known increase in cardiovascular risk in patients treated with COX-2 inhibitors (21, 22).

Our laboratory also employed a number of approaches to enhance expression of 27-hydroxylase and ABCA1. We discovered that these proteins can be upregulated via activation of the adenosine A2A receptor with specific agonists including CGS-21680 and MRE-0094. Ligation of this receptor also inhibits macrophage foam cell transformation under cholesterol loading conditions (20). Since MTX is known to affect both adenosine release and cardiovascular risk, we investigated whether MTX modulates cholesterol metabolism and vulnerability to foam cell formation. We report here that MTX treatment counteracts propensity toward cholesterol overload in THP-1 monocytes/macrophages exposed to IFN-γ or selective COX-2 inhibition. This data supports the hypothesis that MTX provides protection from atherosclerotic cardiovascular disease (ASCVD) by increasing expression of anti-atherogenic molecules involved in cholesterol efflux, likely via a pathway involving adenosine release.

Materials and methods

Cells and reagents

THP-1 monocytes were obtained from ATCC (Manassas, VA).

Oil red O and OptiPrep Density Gradient Media were purchased from Sigma-Aldrich (St. Louis. MO).

Trizol reagent was purchased from Invitrogen (Grand Island, NY).

All reverse transcription- Polymerase chain reaction (RT-PCR) reagents were purchased from Applied Biosystems (Chicago, IL).

Recombinant human IFN-γ was purchased from R&D Systems (Minneapolis, MN).

NS-398 was purchased from RBI-Sigma (Natick, MA).

MTX was purchased from Bedford Laboratories (Bedford Ohio).

Acetylated LDL was purchased from Intracel (Issaquah, Washington).

Anti-cholesterol 27-hydroxylase antibody is an affinity-purified rabbit polyclonal anti-peptide antibody raised against residues 15-28 of the cholesterol 27-hydoxylase protein (23).

Cell culture

THP-1 monocytes were grown at 37°C in a 5% CO2 atmosphere to a density of 106 cells/ml. Growth medium for THP-1 cells was RPMI 1640 (GIBCO BRL, Grand Island, NY) supplemented with 10% Fetal Bovine Serum (FBS) (GIBCO BRL), 50 units/ml penicillin, and 50 units/ml streptomycin. To facilitate differentiation into macrophages, THP-1 monocytes (106 cells/ml) in 12 well plates were treated with 100nM PMA (Sigma) for 4 days at 37°C.

PBMC Isolation

Blood from healthy donors was collected in EDTA treated tubes, pooled and kept at 4°C. The pooled blood was adjusted to a density of 1.120 g/ml with the addition of OptiPrep Density Gradient Media (Sigma) according to the manufacturer's instructions. The blood was then overlaid with a 1.074 g/ml density solution composed of complete RPMI containing 10% FBS and OptiPrep media. A layer of complete RPMI containing 10% FBS was then overlaid on top to prevent monocytes from sticking to the plastic tube. The blood was centrifuged at 750g for 30 minutes at 4°C. After centrifugation, the monocyte interphase was collected from between the 1.074 g/ml and RPMI layer. The collected cells were diluted with 2 volumes of complete RPMI and harvested by centrifugation. The pellet was re-suspended in complete RPMI. The monocytes were counted by hemocytometer and plated at a density of 2×106 cells/well in a 6-well plate.

Experimental conditions

When THP-1 cells had reached 106 cells/ml, media was aspirated and cells were rinsed twice with Dulbecco's Phosphate Buffered Saline (DPBS) without calcium and magnesium. The monocytes were then incubated for 24-48 hr in six well plates, (37°C, 5% CO2) under the following conditions: a) RPMI control; b) RPMI containing 5μM MTX c) RPMI containing NS398 (50 μM); d) RPMI containing NS398 (50 μM) and MTX (increasing doses of 0.1μM, 0.5μM and 5μM); e) RPMI containing IFN-γ (500 U/ml); f) RPMI containing IFN-γ (500 U/ml) and 5μM MTX.

THP-1 macrophages were exposed to the following conditions: a) RPMI control; b) RPMI containing ZM-241385 (10μM); c) RPMI containing MTX (5μM); d) RPMI containing IFN-γ (500 U/ml); e) RPMI containing IFN-γ (500 U/ml) and 5μM MTX; f) RPMI containing ZM-241385 (10μM) and MTX (5μM); g) RPMI containing IFN-γ (500 U/ml), ZM-241385 (10μM) and MTX (5μM); h) RPMI containing NS398 (50μM), ZM-241385 (10μM) and MTX (5μM).

Immediately after the incubation period, the cells were collected and centrifuged at 1500 RPM at room temperature, media was aspirated and cell protein and RNA were isolated.

PBMC were incubated for 18 h in RPMI with 10% FBS with and without the addition of MTX at a concentration of 5μM. Cells were collected and RNA isolated.

RNA isolation

RNA was isolated using 1ml Trizol reagent per 106 cells and dissolved in nuclease-free water. The quantity of total RNA from each condition was measured by absorption at 260 and 280 wavelengths using quartz cuvettes by ultraviolet spectrophotometry (Hitachi U2010 spectrophotometer).

Analysis of 27-hydroxylase message by RT-PCR

27-hydroxylase and ABCA1 mRNA were quantitated by real-time PCR. cDNA was copied from 5 μg of total RNA using M-MLV reverse transcriptase primed with oligo dT. Equal amounts of cDNA were taken from each RT reaction mixture for PCR amplification using cholesterol 27-hydroxylase-specific primers or ABCA1 specific primers as well as glyceraldehyde-3-phosphate dehydrogenase (GAPDH) control primers. The cholesterol 27-hydroxylase-specific primers span a 311 base-pair sequence encompassing nucleotides 491-802 of the human cholesterol 27-hydroxylase cDNA (24). ABCA1 primers yield a 234 BP amplified fragment (21). Real-time PCR analysis was performed using the SYBR Green PCR Reagents Kit (Applied Biosystems) with a Stratagene MX3005P QPCR System.

PCR was performed using techniques standardized in our laboratory. Each PCR reaction contained 2.5 μl of the 10x fluorescent green buffer, 3 μl of 25 mM MgCl2, 2 μl dNTP mix (2500 μM dCTP, 2500 μM dGTP, 2500 μM dATP, and 5000 μM dUTP), 0.15 μl polymerase (5 U/μl; AmpliTaq Gold; Applied Biosystems), 0.25 μl uracil-N-glycosylase (1 U/μl UNG; AmpErase; Applied Biosystems), 0.5 μl of the forward and reverse primers (10μM concentration), 4 μl cDNA, and water to a final volume of 25 μl. The thermal cycling parameters were as follows: 5 min at 95°C to activate the polymerase (AmpliTaq Gold; Applied Biosystems), followed by 45 cycles of 30 seconds at 95°C and 45 seconds at 58°C then 45 seconds at 72°C. Each reaction was done in triplicate. The amounts of PCR products were estimated, using software provided by the manufacturer (Stratagene). After completion of PCR cycles, the reactions were heat denatured over a 35°C temperature gradient from 60°C to 95°C. To correct for differences in cDNA load among samples, the target PCRs were normalized to a reference PCR involving the endogenous housekeeping genes glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and β-actin. Nontemplate controls were included for each primer pair to check for significant levels of any contaminants. Fluorescence emission spectra were monitored and analyzed. PCR products were measured by the threshold cycles (CT), at which specific fluorescence becomes detectable. The CT was used for kinetic analysis and was proportional to the initial number of target quantity copies in the sample. A melting curve analysis was performed to assess the specificity of the amplified PCR products. The quantity of the samples was calculated after the CTs of the serial dilutions were compared with a control. QRT-PCR standards were prepared by making 1:10 serial dilutions of a purified PCR product.

Protein extraction and Western blot analysis

Total cell lysates were prepared for Western immunoblotting using RIPA lysis buffer (98% PBS, 1% Igepal CA-630, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate [SDS]). 100μl of RIPA lysis buffer and 10μl of protease inhibitor cocktail (Sigma) were added to the cell pellet from each condition and incubated on ice for 35 min with vortexing every 5min. Supernatants were collected after centrifuging at 10,000g at 4°C for 10 min using an Eppendorf 5415C centrifuge. The quantity of protein in each supernatant was measured by absorption at 560nm using a Hitachi U2010 spectrophotometer.

Total cell lysate was used for Western blots. Protein samples (20μg/lane) were boiled for 5 min, loaded onto a 10% polyacrylamide gel, electrophoresed for 1.5 hr at 100V then transferred to a nitrocellulose membrane in a semi-dry transblot apparatus for 1 hr at 100V. The nitrocellulose membrane was blocked for 4 hr at 4°C in blocking solution (3% nonfat dry milk dissolved in 1×Tween20-tris-buffered saline [TTBS]) then immersed in a 1:300 dilution of primary antibody (18.7 μg/ml) in blocking solution overnight at 4°C. The primary antibody is an affinity-purified rabbit polyclonal anti-peptide antibody raised against residues 15-28 of the cholesterol 27-hydroxylase protein (23). The following day, the membrane was washed 5 times in TTBS for 5mins per wash then incubated at room temperature in a 1:3000 dilution of ECL donkey anti-rabbit IgG Horseradish peroxidase-linked species-specific whole antibody (Amersham Biosciences, product Code NA934). The five washes in TTBS were repeated, then the immunoreactive protein was detected using ECL western blotting detection reagent (Amersham Biosciences, Cat No RPN2106) and film development in SRX-101A (Konica Minolta).

As control, on the same transferred membrane, beta-actin was detected using mouse anti-beta-actin (diluted in 1:1000, from abCam, product Code: ab6276) and ECL sheep anti-mouse-IgG Horseradish peroxidase-linked species-specific whole antibody (diluted in 1:2000, from Amersham Biosciences, product Code NA931) and all other similar steps as above.

For ABCA1 detection, macrophage cell lysates were electrophoresed for 1.5 hr at 100V (10% polyacrylamide gel), then transferred to a nitrocellulose membrane. The membrane was blocked for 4 hr at 4°C in blocking solution then incubated overnight at 4°C in a 1:200 dilution of rabbit anti- ABCA1antibody (Santa Cruz Biotechnology). The following day, the membrane was washed 5 times in TTBS for 5mins per wash then incubated at room temperature in a 1:5000 dilution of ECL donkey anti-rabbit IgG Horseradish peroxidase-linked species-specific whole antibody. Development proceeded as described above for the 27-hydroxylase antibody.

Foam cell formation and staining

THP-1 differentiated macrophages were washed three times with phosphate-buffered saline (PBS) and further incubated in RPMI (37°C, 5% CO2) for 48 hr under the following five conditions: a) acetylated LDL (50 μg/ml); b) acetylated LDL (50μg/ml) and IFN-γ (500U/ml); c) acetylated LDL (50μg/ml) and IFN-γ neutralizing antibody (1.2μg/ml); d) acetylated LDL (50μg/ml), IFN-γ (500U/ml) and IFN-γ neutralizing antibody (1.2μg/ml); e) acetylated LDL (50μg/ml) and IFN-γ receptor antibody (125ng/ml); f) acetylated LDL (50μg/ml), IFN-γ receptor antibody (125ng/ml) and IFN-γ (500U/ml).

Immediately following incubation, media was aspirated and cells were fixed in the same 12 well plates used for incubation, with 4% paraformaldehyde in water, for 2-4 min. Cells were stained with 0.2% Oil-Red-O in methanol for 1-3 min. Cells were observed via light microscope (Axiovert 25-Zeiss) with 100X magnification and then photographed using a Kodak DC 290 Zoom Digital Camera. The number of foam cells formed in each condition were calculated manually and presented as percentage foam cell formation.

Data Analysis

Statistical analysis were performed using GraphPad version 4.02 (GraphPad, San Diego, CA). All data were analyzed by one-way ANOVA and pairwise multiple comparisions were made between control and treatment conditions using Bonferroni's method.

Results

MTX Increases 27-Hydroxylase Message and Blocks COX-2 Inhibitor and IFN-γ -mediated 27-Hydroxylase Downregulation in THP-1 Monocytes

MTX (5μM, 18 hr) increased 27-hydroxylase mRNA expression (113.9±6.4%) and completely blocked NS398-induced downregulation of 27-hydroxylase message (112.8±13.1% for NS398+MTX versus 71.1±4.3% for NS398 alone, with untreated as 100%, n=3, p<0.01) (Figure 1A). MTX (5μM, 18 hr) also abrogated IFN-γ-induced downregulation of 27-hydroxylase message (86± 9.6% of control for IFN-γ+MTX versus 45± 6.0% for IFN-γ alone, with untreated as 100%, n=3, p=0.02) (Figure 1B).

Figure 1. Effect of MTX on 27-hydroxylase message in THP-1 monocytes and human PBMC.

Figure 1

(a) Quantitation of 27-hydroxylase message in NS-398-treated THP-1 cells exposed to MTX. COX-2 inhibitor-mediated decrease in 27-hydroxylase mRNA is prevented by MTX. THP-1 human monocytes were exposed to the following conditions represented by the four bars (from left to right): (1) Control RPMI 1640, (2) MTX (5 μM, 18 hr), (3) NS398 (50μM, 18 hr), (4) MTX (5 μM, 18 hr) and NS398 (50μM, 18hr). Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA expression by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. *p<0.05, control vs. NS398. #p<0.01, NS398 + MTX vs. NS398. (b) Quantitation of 27-hydroxylase message in IFN-γ-treated THP-1 cells exposed to MTX. IFN-γ-mediated decrease in 27-hydroxylase mRNA is prevented by MTX. THP-1 human monocytes were exposed to the following conditions represented by the three bars (from left to right): (1) Control RPMI 1640, (2) IFN-γ (500 U/ml, 18 hr), (3) IFN-γ (500 U/ml, 18 hr) and MTX (5 μM, 18 hr). Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA expression by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. ***p=0.02 IFN-γ + MTX vs IFN-γ. (c) Effect of MTX on 27-hydroxylase message in healthy donor PBMC. Isolated PBMCs were incubated in RPMI 1640 with and without MTX (5 μM, 18 hr). Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA expression by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. ****p=0.004 MTX vs control untreated.

This ability of MTX to overcome suppression of 27-hydroxylase expression by NS-398 was observed at MTX doses of 0.1μM, 0.5μM and 5μM at both the protein (Figure 2A) and message level (Figure 2B).

Figure 2. Detection and quantitation of cholesterol 27-hydroxylase in NS398-treated THP-1 cells exposed to increasing doses of MTX.

Figure 2

(a) Decrease in 27-hydroxylase protein in THP-1 monocytes treated with the COX-2 inhibitor NS398 is corrected with increasing concentrations of MTX. Cultured THP-1 monocytic cells were untreated or exposed to NS398 (50μM, 18hr) then untreated or exposed to increasing doses of MTX for 24 hr. Total cell protein was isolated and 27-hydroxylase detected with specific rabbit polyclonal anti-human 27-hydroxylase antibody. Western blotting was also performed with an anti-beta actin antibody to confirm equal protein loading. (b) COX-2-inhibitor-mediated suppression of 27-hydroxylase mRNA expression in THP-1 monocytes is overcome with MTX. Cultured THP-1 monocytic cells were incubated in NS398 (50μM, 48hr) then untreated or exposed to increasing doses of MTX for 24 hr. Following isolation of total RNA, the RNA was reverse transcribed and the cDNA amplified by QRT-PCR as described. Signals obtained from the amplification of GAPDH message were used as internal controls. *p<0.05, **p<0.01, MTX vs Control (C). #p<0.01, NS+MTX vs NS398 (NS).

Effect of MTX on 27-Hydroxylase Message in Human Donor PBMC

To make certain that our THP-1 model accurately represents primary human monocyte behavior, we isolated PBMC from healthy human donors and incubated them for 18h in RPMI 1640 media in the presence and absence of MTX (5μM). MTX exposure resulted in a nearly fourfold increase in 27-hydroxylase mRNA (3.63±0.46 for MTX with untreated as 1.00, n=3, p=0.004) as assessed by QRT-PCR (Figure 1C).

MTX Reverses COX-2 Inhibitor-Induced and INF-γ-Induced Downregulation of ABCA1 and 27-Hydroxylase via Adenosine A2A Receptor Activation

MTX was also effective in blocking COX-2 inhibitor-mediated downregulation of ABCA1 message in THP-1 monocytes. Adenosine A2A receptor blockade with ZM-241385 abolished the ability of MTX to counter COX-2 inhibitor effects on both 27-hydroxylase (Figure 3A).and ABCA1 (Figure 3B).

Figure 3. Detection and quantitation of cholesterol 27-hydroxylase and ABCA1 mRNA in NS398-treated THP-1 cells exposed to MTX in the presence and absence of A2A receptor antagonism with ZM-241385.

Figure 3

(a) Suppression of 27-hydroxylase message in THP-1 cells by NS398 is reversed by MTX and this reversal is blocked by ZM-241385. THP-1 monocytes were exposed to the following conditions represented by the four bars from left to right: (1) Control RPMI 1640, (2) NS398 (50μM, 24 hr), (3) NS398 (50μM, 24 hr) then add MTX (5 μM, 24 hr), (4) NS398 (50μM) and ZM-241385 (10μM) for 24 hr, then add MTX (5μM) for 24 hr. Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. (b) Suppression of ABCA1 message in THP-1 cells by NS398 is reversed by MTX and this reversal is blocked by ZM-241385. THP-1 monocytes were exposed to the following conditions represented by the four bars from left to right: (1) Control RPMI 1640, (2) NS398 (50μM, 24 hr), (3) NS398 (50μM, 24 hr) then add MTX (5 μM, 24 hr), (4) NS398 (50μM) and ZM-241385 (10μM) for 24 hr, then add MTX (5μM) for 24 hr. Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. *p<0.05, **p<0.01, MTX vs control (C). #p<0.01, NS398+MTX vs NS398.

Similarly, downregulation of 27-hydroxylase and ABCA1 by IFN-γ, which we showed previously in THP-1 monocytes (19), was also demonstrated in THP-1 macrophages and was prevented by MTX and this effect of MTX was negated by ZM-241385 (Figure 4A-C). THP-1 macrophages exposed to MTX alone exhibited a substantial increase in expression of both 27-hydroxylase and ABCA1 relative to untreated THP-1 macrophages (Figure 4A-C).

Figure 4. Detection and quantitation of cholesterol 27-hydroxylase mRNA and protein and ABCA1 mRNA in IFN-γ-stimulated THP-1 macrophages exposed to MTX in the presence and absence of A2A receptor antagonism with ZM-241385.

Figure 4

(a) Suppression of 27-hydroxylase message in THP-1 macrophages by IFN-γ is reversed by MTX and this reversal is blocked by ZM-241385. THP-1 macrophages were exposed to the following conditions represented by the eight bars from left to right: (1) Control RPMI 1640, (2) ZM-241385 (10μM, 24 hr), (3) MTX (5 μM, 24 hr), (4) IFN-γ (500 U/ml, 24 hr), (5) IFN-γ (500 U/ml, 24 hr), then add MTX (5μM, 24 hr), (6) ZM-241385 (10 μM, 24 hr), then add MTX (5μM, 24 hr), (7) ZM-241385 (10 μM) and IFN-γ (500 U/ml) for 24 hr, then add MTX (5μM, 24 hr), (8) ZM-241385 (10 μM) and NS398 (50μM) for 24 hr, then add MTX (5μM, 24 hr). Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. (b) Suppression of 27-hydroxylase protein in THP-1 cells by IFN-γ is reversed by MTX and this reversal is blocked by ZM-241385. THP-1 macrophages were exposed to identical conditions 1-8 as in part (a) of this figure represented by the eight lanes of the immunoblot from left to right. Total cell protein was isolated and 27-hydroxylase detected with specific rabbit polyclonal anti-human 27-hydroxylase antibody. Western blotting was also performed with an anti-beta actin antibody to confirm equal protein loading. (c) Suppression of ABCA1 message in THP-1 macrophages by IFN-γ is reversed by MTX and this reversal is blocked by ZM-241385. THP-1 macrophages were exposed to identical conditions 1-8 as in part (a) of this figure represented by the eight bars from left to right. Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. **p<0.01, IFN-γ+ MTX vs IFN-γ. #p<0.01, ZM+MTX vs MTX.

Adenosine A2A Receptor Activation Increases 27-Hydroxylase and Blocks COX-2 Inhibitor-mediated 27-Hydroxylase Downregulation

Addition of the adenosine A2A receptor agonist CGS-21680 to THP-1 monocytes exposed to NS398 overcame the reduction in 27-hydroxylase expression. This is demonstrated by immunoblot (Figure 5A) and QRT-PCR (Figure 5B). Addition of CGS-21680 to NS398-treated THP-1 cells gave rise to a 184% increase in 27-hydroxylase mRNA (167.2±8.57% in CGS+NS398 vs. 58.9±2.3% in NS398 alone, n=3, p<0.001) (Figure 5B).

Figure 5. Effect of the A2AR agonist CGS0-21680 on NS398-induced suppression of 27-hydroxylase expression in THP-1 monocytes.

Figure 5

a) 27-hydroxylase message level is decreased by the COX-2 inhibitor, NS398 (50μM), and this decrease is reversed by the addition of the adenosine A2AR agonist, CGS-21680 (10 μM). THP-1 monocytes were exposed to the following conditions represented by the four bars from left to right: (1) Control RPMI 1640, (2) CGS-21680 (10μM, 18 hr), (3) NS398 (50 μM, 18 hr), (4) NS398 (50μM, 18 hr) and CGS-21680 (10 μM, 18 hr). Cells were extracted for total RNA, and evaluated for 27-hydroxylase mRNA by QRT-PCR. Signals obtained from the amplification of GAPDH message were used as internal controls. b) 27-hydroxylase protein level is decreased by the COX-2 inhibitor, NS398, and this decrease is reversed by the addition of the adenosine A2AR agonist, CGS-21680. THP-1 monocytes were exposed to the following conditions represented by the four bands from left to right: (1) Control RPMI 1640, (2) CGS-21680 (10μM, 18 hr), (3) NS398 (50 μM, 18 hr), (4) NS398 (50μM, 18 hr) and CGS-21680 (10 μM, 18 hr). *p<0.01, control vs. NS398. #p<0.01, NS398 + CGS-21680 vs. NS398.

MTX Attenuates Foam Cell Transformation in Lipid-loaded THP-1 Macrophages

Acetylated LDL-treated THP-1 macrophages showed a significant decrease in foam cell transformation in the presence of MTX compared to control (29.7±2.0% vs. 39.3±5.0%, p<0.001) (Figure 6A). NS398 treatment resulted in 72.7±4.9% foam cells while combined NS398+MTX resulted in only 36.3±3.2% foam cells, (n=3, p<0.001) (Figure 6B). IFN-γ treatment prior to cholesterol loading with acetylated LDL resulted in 71.0±5.0% foam cells while IFN-γ+MTX resulted in only 46.0±7.2% foam cells,(n=3, p<0.001) (Figure 6C). Preincubation of THP-1 macrophages with the selective A2A receptor antagonist (ZM-241385) prior to MTX treatment ablated the anti-atherogenic effect of MTX and resulted in a significant increase in foam cells (62.1 ±1.5%) (Figure 6D).

Figure 6. Effect of MTX on NS398 and IFN-γ-induced foam cell transformation in lipid loaded THP-1 macrophages.

Figure 6

Representative photomicrographs at 40 X magnification of lipid laden macrophages stained with oil red-O. A) Acetylated LDL-treated THP-1 macrophages showed a significant decrease in foam cell transformation in the presence of MTX compared to control B) MTX prevented the NS398-induced increase in foam cell formation in THP-1 macrophages. C) MTX prevented the IFN-γ-induced increase in foam cell formation in THP-1 macrophages. D) Effectiveness of MTX in decreasing foam cell formation is abolished by A2AR antagonism with ZM-241385

Discussion

MTX modulates the expression of numerous inflammatory cytokines. Methotrexate has been successfully used in the treatment of many immune- or inflammatory-mediated diseases (eg, rheumatoid arthritis) by causing generalized immunomodulation. Several studies have demonstrated that binding of adenosine to the A2 receptor inhibits lymphocyte proliferation and production of TNF, IL-8, and IL-12, as well as increases secretion of IL-10 (25). Given the important role of inflammatory mediators in the pathogenesis of ASCVD, therapeutic modulation targeting inflammatory mediators might be a new and promising strategy for treating ASCVD.

Prior studies demonstrate that adenosine A2A receptor ligation can help prevent atherosclerosis in an experimental intimal injury model (26). In this model the recruitment of inflammatory cells to the intima following endothelial injury was markedly reduced. Our results suggest that MTX, which increases adenosine levels, may have a similar effect. Moreover, our results further suggest that MTX, and adenosine, might reduce atherosclerosis not only by reducing inflammation in the vessel wall but also by stimulating reverse cholesterol transport.

The concentration of MTX used was in the range of prior in vitro cell culture studies (27). In our experiments, the concentration of MTX (5 mM) falls within the span of serum concentrations described in the literature (28-31). Although previously measured serum MTX levels extend through a wide range (0.5-1800 μM), the relevance of serum levels to MTX effects is unclear since antiinflammatory activity has been attributed to multiple metabolites of methotrexate rather than methotrexate itself, some of which have vastly extended tissue half-lives (32).

Recent studies by Ghosh and colleagues (33) suggest that another mechanism by which COX-2 promotes atherosclerotic cardiovascular disease is by diminishing COX-2-mediated metabolism of endocannabinoids and downstream activation of PPARδ increasing endothelial tissue factor expression. Tissue factor promotes intravascular thrombosis and the increased expression of tissue factor likely predisposes to the further development of atherosclerotic cardiovascular disease. MTX may also reduce tissue factor expression by increasing adenosine levels and adenosine A2A receptor activation since it has been known for some time that adenosine suppresses tissue factor production (34-36).

In studies published recently, 27-hydroxycholesterol was shown to antagonize estrogen at its receptors, an effect which may negatively impact the development of atherosclerosis (37, 38). Although this effect may be an important factor in preventing postmenopausal estrogen therapy from diminishing the risk of atherosclerosis, it may not be relevant to the development of atherosclerosis in males. Moreover, conjugated estrogens also reduce blood concentrations of such anticoagulants as plasminogen activator inhibitor-1 (PAI-1) (39) and rising 27-hydroxycholesterol levels may reverse these estrogen-mediated effects. Furthermore, anti-atherogenic rather than pro-atherogenic effects have been observed with selective estrogen receptor modulators (SERMs) such as raloxifene and tamoxifen in vivo. Potential explanations have included favourable alterations of anti-oxidant activity, cholesterol, fibrinogen and homocysteine levels as well as vascular tone (40). Thus, the overall impact of possible estrogen receptor blockade on atherogenicity by 27-hydroxycholesterol may be eclipsed by a multitude of other factors at play.

This is the first evidence that any widely used pharmacotherapy can increase the expression of the anti-atherogenic 27-hydroxylase or ABCA1 and can counteract the effects of COX-2 inhibition or IFN-γ exposure on gene expression. We have demonstrated that MTX inhibits foam cell formation under conditions of lipid overload. Our results suggest that the capacity of MTX to reduce the burden of ASCVD in patients with RA may be ascribed, in part, to favorable alterations in cholesterol homeostasis mediated via activation of the adenosine A2A receptor. Thus, adenosine receptor ligation may provide a suitable mechanism for the development of a promising treatment paradigm with long term-benefit in ASCVD.

Acknowledgements

This work was supported by a grant from the National Institutes of Health/National Heart, Lung and Blood Institute HL073814 (Reiss). Additional support was provided by the Arthritis National Research Foundation (Chan). National Institutes of Health (AA13336, AR41911 and GM56268), King Pharmaceuticals, the General Clinical Research Center (M01RR00096) and by the Kaplan Cancer Center of New York University School of Medicine (Cronstein).

References

  • 1.Ward JR. Historical perspective on the use of methotrexate for the treatment of rheumatoid arthritis. J Rheumatol. 1985;12(suppl 12):3–6. [PubMed] [Google Scholar]
  • 2.Kremer JM. Historical overview of the treatment of rheumatoid arthritis with an emphasis on methotrexate. J Rheumatol. 1996;44:34–7. [PubMed] [Google Scholar]
  • 3.Alarcon GS. Methotrexate use in rheumatoid arthritis. A clinician's perspective. Immunopharmacology. 2000;47:259–71. doi: 10.1016/s0162-3109(00)00184-3. [DOI] [PubMed] [Google Scholar]
  • 4.Moss RB. Alternative pharmacotherapies for steroid-dependent asthma. Chest. 1995;107:817–25. doi: 10.1378/chest.107.3.817. [DOI] [PubMed] [Google Scholar]
  • 5.Wong JM, Esdaile JM. Methotrexate in systemic lupus erythematosus. Lupus. 2005;14:101–5. doi: 10.1191/0961203305lu2043rr. [DOI] [PubMed] [Google Scholar]
  • 6.Feagan BG, Rochon J, Fedorak RN, Irvine EJ, Wild G, Sutherland L, et al. Methotrexate for the treatment of Crohn's disease. The North American Crohn's Study Group Investigators. N Engl J Med. 1995;332:292–7. doi: 10.1056/NEJM199502023320503. [DOI] [PubMed] [Google Scholar]
  • 7.Carlson JA, Cavaliere LF, Grant-Kels JM. Cutaneous vasculitis: diagnosis and management. Clin Dermatol. 2006;24:414–29. doi: 10.1016/j.clindermatol.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 8.Ytterberg SR. Treatment of refractory polymyositis and dermatomyositis. Curr Rheumatol Rep. 2006;8:167–73. doi: 10.1007/s11926-996-0021-7. [DOI] [PubMed] [Google Scholar]
  • 9.Hornung N, Stengaard-Pedersen K, Ehrnrooth E, Ellingsen T, Poulsen JH. The effects of low-dose methotrexate on thymidylate synthase activity in human peripheral blood mononuclear cells. Clin Exp Rheumatol. 2000;18:691–8. [PubMed] [Google Scholar]
  • 10.Morabito L, Montesinos MC, Schreibman DM, Balter L, Thompson LF, Resta R, Carlin G, Huie MA, Cronstein BN. Methotrexate and sulfasalazine promote adenosine release by a mechanism that requires ecto-5′-nucleotidase-mediated conversion of adenine nucleotides. J Clin Invest. 1998;101:295–300. doi: 10.1172/JCI1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Xu Z, Mueller RA, Park SS, Boysen PG, Cohen MV, Downey JM. Cardioprotection with adenosine A2 receptor activation at reperfusion. J Cardiovasc Pharmacol. 2005;46:794–802. doi: 10.1097/01.fjc.0000188161.57018.29. [DOI] [PubMed] [Google Scholar]
  • 12.Sitkovsky MV, Lukashev D, Apasov S, Kojima H, Koshiba M, Caldwell C, Ohta A, Thiel M. Physiological control of immune response and inflammatory tissue damage by hypoxia-inducible factors and adenosine A2A receptors. Annu Rev Immunol. 2004;22:657–82. doi: 10.1146/annurev.immunol.22.012703.104731. [DOI] [PubMed] [Google Scholar]
  • 13.Gonzalez A, Maradit Kremers H, Crowson CS, Ballman KV, Roger VL, Jacobsen SJ, O'fallon WM, Gabriel SE. Do cardiovascular risk factors confer the same risk for cardiovascular outcomes in rheumatoid arthritis patients as in non-RA patients? Ann Rheum Dis. 2007 May 22; doi: 10.1136/ard.2006.059980. [DOI] [PubMed] [Google Scholar]
  • 14.Gonzalez-Gay MA, Gonzalez-Juanatey C, Miranda-Filloy JA, Garcia-Porrua C, Llorca J, Martin J. Cardiovascular disease in rheumatoid arthritis. Biomed Pharmacother. 2006;60:673–7. doi: 10.1016/j.biopha.2006.09.006. [DOI] [PubMed] [Google Scholar]
  • 15.Kremers HM, Gabriel SE. Rheumatoid arthritis and the heart. Curr Heart Fail Rep. 2006;3:57–63. doi: 10.1007/s11897-006-0003-0. [DOI] [PubMed] [Google Scholar]
  • 16.Solomon DH, Curhan GC, Rimm EB, Cannuscio CC, Karlson EW. Cardiovascular risk factors in women with and without rheumatoid arthritis. Arthritis Rheum. 2004;50:3444–49. doi: 10.1002/art.20636. [DOI] [PubMed] [Google Scholar]
  • 17.van Halm VP, Nurmohamed MT, Twisk JW, Dijkmans BA, Voskuyl AE. Disease-modifying antirheumatic drugs are associated with a reduced risk for cardiovascular disease in patients with rheumatoid arthritis: a case control study. Arthritis Res Ther. 2006;8:R151. doi: 10.1186/ar2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Choi HK, Hernan MA, Seeger JD, Robins JM, Wolfe F. Methotrexate and mortality in patients with rheumatoid arthritis: a prospective study. Lancet. 2002;359:1173–77. doi: 10.1016/S0140-6736(02)08213-2. [DOI] [PubMed] [Google Scholar]
  • 19.Reiss AB, Awadallah NW, Malhotra S, Montesinos MC, Chan ESL, Javitt NB, Cronstein BN. Immune complexes and interferon-γ decrease cholesterol 27-hydroxylase in human arterial endothelium and macrophages. J Lipid Res. 2001;42:1913–22. [PubMed] [Google Scholar]
  • 20.Reiss AB, Rahman MM, Chan ESL, 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. J Leukoc Biology. 2004;76:727–34. doi: 10.1189/jlb.0204107. [DOI] [PubMed] [Google Scholar]
  • 21.Chan ES, Zhang H, Fernandez P, Edelman SD, Pillinger MH, Ragolia L, Palaia T, Carsons SE, Reiss AB. Effect of COX Inhibition on Cholesterol Efflux Proteins and Atheromatous Foam Cell Transformation in THP-1 Human Macrophages: A Possible Mechanism for Increased Cardiovascular Risk. Arthritis Res Ther. 2007;9:R4. doi: 10.1186/ar2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Iezzi A, Ferri C, Mezzetti A, Cipollone F. COX-2: Friend or Foe? Curr Pharm Des. 2007;13:1715–21. doi: 10.2174/138161207780831293. [DOI] [PubMed] [Google Scholar]
  • 23.Cali JJ, Russell DW. Characterization of human sterol 27-hydroxylase. J Biol Chem. 1991;266:7774–78. [PubMed] [Google Scholar]
  • 24.Reiss AB, Martin KO, Rojer DE, Iyer S, Grossi EA, Galloway AC, Javitt NB. Sterol 27-hydroxylase: expression in human arterial endothelium. J Lipid Res. 1997;38:1254–60. [PubMed] [Google Scholar]
  • 25.Cutolo M, Sulli A, Pizzorni C, Seriolo B, Straub RH. Anti-inflammatory mechanisms of methotrexate in rheumatoid arthritis. Ann Rheum Dis. 2001;60:729–35. doi: 10.1136/ard.60.8.729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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. Arterioscler Thromb Vasc Biol. 2001;21:791–96. doi: 10.1161/01.atv.21.5.791. [DOI] [PubMed] [Google Scholar]
  • 27.Galivan J, Pupons A, Rhee MS. Hepatic parenchymal cell glutamylation of methotrexate studied in monolayer culture. Cancer Res. 1986;46:670–5. [PubMed] [Google Scholar]
  • 28.Cheng KK. Association of plasma methotrexate, neutropenia, hepatic dysfunction, nausea/vomiting and oral mucositis in children with cancer. Eur J Cancer Care (Engl) 2008;17:306–11. doi: 10.1111/j.1365-2354.2007.00843.x. [DOI] [PubMed] [Google Scholar]
  • 29.Bacci G, Loro L, Longhi A, Bertoni F, Bacchini P, Versari M, Picci P, Serra M. No correlation between methotrexate serum level and histologic response in the pre-operative treatment of extremity osteosarcoma. Anticancer Drugs. 2006;17:411–15. doi: 10.1097/01.cad.0000203379.14738.d9. [DOI] [PubMed] [Google Scholar]
  • 30.Wallace CA, Bleyer WA, Sherry DD, Salmonson KL, Wedgwood RJ. Toxicity and serum levels of methotrexate in children with juvenile rheumatoid arthritis. Arthritis Rheum. 1989;32:677–81. doi: 10.1002/anr.1780320604. [DOI] [PubMed] [Google Scholar]
  • 31.Ravelli A, Di Fuccia G, Molinaro M, Ramenghi B, Zonta L, Regazzi MB, Martini A. Plasma levels after oral methotrexate in children with juvenile rheumatoid arthritis. J Rheumatol. 1993;20:1573–7. [PubMed] [Google Scholar]
  • 32.Chan ES, Cronstein BN. Molecular action of methotrexate in inflammatory diseases. Arthritis Res. 2002;4:266–73. doi: 10.1186/ar419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ghosh M, Wang H, Ai Y, Romeo E, Luyendyk JP, Peters JM, Mackman N, Dey SK, Hla T. COX-2 suppresses tissue factor expression via endocannabinoid-directed PPAR{delta} activation. J Exp Med. 2007;204:2053–61. doi: 10.1084/jem.20070828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Deguchi H, Takeya H, Wada H, Gabazza EC, Hayashi N, Urano H, Suzuki K. Dilazep, an antiplatelet agent, inhibits tissue factor expression in endothelial cells and monocytes. Blood. 1997;90:2345–56. [PubMed] [Google Scholar]
  • 35.Deguchi H, Takeya H, Urano H, Gabazza EC, Zhou H, Suzuki K. Adenosine regulates tissue factor expression on endothelial cells. Thromb Res. 1998;91:57–64. doi: 10.1016/s0049-3848(98)00045-0. [DOI] [PubMed] [Google Scholar]
  • 36.Watanabe T, Tokuyama S, Yasuda M, Sasaki T, Yamamoto T. Involvement of adenosine A2 receptors in the changes of tissue factor-dependent coagulant activity induced by polymorphonuclear leukocytes in endothelial cells. Jpn J Pharmacol. 2002;88:407–13. doi: 10.1254/jjp.88.407. [DOI] [PubMed] [Google Scholar]
  • 37.Umetani M, Domoto H, Gormley AK, Yuhanna IS, Cummins CL, Javitt NB, Korach KS, Shaul PW, Mangelsdorf DJ. 27-Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nat Med. 2007 Sep 16; doi: 10.1038/nm1641. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 38.Dusell CD, Umetani M, Shaul PW, Mangelsdorf DJ, McDonnell DP. 27-Hydroxycholesterol is an endogenous Selective Estrogen Receptor Modulator (SERM) Mol Endocrinol. 2007 Sep 13; doi: 10.1210/me.2007-0383. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Skouby SO, Sidelmann JJ, Nilas L, Jespersen J. A comparative study of the effect of continuous combined conjugated equine estrogen plus medroxyprogesterone acetate and tibolone on blood coagulability. Hum Reprod. 2007;22:1186–91. doi: 10.1093/humrep/del498. [DOI] [PubMed] [Google Scholar]
  • 40.Leung FP, Tsang SY, Wong CM, Yung LM, Chan YC, Leung HS, Yao X, Huang Y. Raloxifene, tamoxifen and vascular tone. Clin Exp Pharmacol Physiol. 2007;34(8):809–13. doi: 10.1111/j.1440-1681.2007.04684.x. [DOI] [PubMed] [Google Scholar]

RESOURCES