Skip to main content

A novel hybrid aspirin-NO-releasing compound inhibits TNFalpha release from LPS-activated human monocytes and macrophages

Abstract

Background

The cytoprotective nature of nitric oxide (NO) led to development of NO-aspirins in the hope of overcoming the gastric side-effects of aspirin. However, the NO moiety gives these hybrids potential for actions further to their aspirin-mediated anti-platelet and anti-inflammatory effects. Having previously shown that novel NO-aspirin hybrids containing a furoxan NO-releasing group have potent anti-platelet effects, here we investigate their anti-inflammatory properties. Here we examine their effects upon TNFα release from lipopolysaccharide (LPS)-stimulated human monocytes and monocyte-derived macrophages and investigate a potential mechanism of action through effects on LPS-stimulated nuclear factor-kappa B (NF-κB) activation.

Methods

Peripheral venous blood was drawn from the antecubital fossa of human volunteers. Mononuclear cells were isolated and cultured. The resultant differentiated macrophages were treated with pharmacologically relevant concentrations of either a furoxan-aspirin (B8, B7; 10 μM), their respective furazan NO-free counterparts (B16, B15; 10 μM), aspirin (10 μM), existing nitroaspirin (NCX4016; 10 μM), an NO donor (DEA/NO; 10 μM) or dexamethasone (1 μM), in the presence and absence of LPS (10 ng/ml; 4 h). Parallel experiments were conducted on undifferentiated fresh monocytes. Supernatants were assessed by specific ELISA for TNFα release and by lactate dehydrogenase (LDH) assay for cell necrosis. To assess NF-κB activation, the effects of the compounds on the loss of cytoplasmic inhibitor of NF-κB, IκBα (assessed by western blotting) and nuclear localisation (assessed by immunofluorescence) of the p65 subunit of NF-κB were determined.

Results

B8 significantly reduced TNFα release from LPS-treated macrophages to 36 ± 10% of the LPS control. B8 and B16 significantly inhibited monocyte TNFα release to 28 ± 5, and 49 ± 9% of control, respectively. The B8 effect was equivalent in magnitude to that of dexamethasone, but was not shared by 10 μM DEA/NO, B7, the furazans, aspirin or NCX4016. LDH assessment revealed none of the treatments caused significant cell lysis. LPS stimulated loss of cytoplasmic IκBα and nuclear translocation of the p65 NF-κB subunit was inhibited by the active NO-furoxans.

Conclusion

Here we show that furoxan-aspirin, B8, significantly reduces TNFα release from both monocytes and macrophages and suggest that inhibition of NF-κB activation is a likely mechanism for the effect. This anti-inflammatory action highlights a further therapeutic potential of drugs of this class.

Background

Aspirin (acetylsalicylic acid) was first synthesized in 1899 and was the first example of the family of nonsteroidal anti-inflammatory drugs (NSAIDs). Its therapeutic uses include the treatment of headache, rheumatic pain and inflammation, in addition to being utilised as an effective prophylactic against thrombotic events in the cardiovascular system. The anti-inflammatory effects of aspirin are achieved primarily through inhibition of cyclooxygenase-mediated synthesis of pro-inflammatory prostanoids [1]; it causes irreversible inhibition by selectively and rapidly acetylating a serine residue (Ser 530) near the C-terminus of the cyclooxygenase (COX) family of enzymes, forming an impediment to the binding of arachidonic acid [2–4]. The acetylation evokes a requirement for new COX to be synthesised for subsequent production of prostaglandins. Unfortunately, gastrointestinal disorders, including ulceration, are a common side-effect of aspirin, limiting its long-term use [5–8]. The effect is primarily believed to be due to inhibition of the production of prostaglandins that normally protect the gastric mucosa [8–11].

Aspirin esters containing a nitric oxide (NO)-donor moiety overcome the gastric side-effects [12, 13] likely via the cytoprotective effects of drug-derived NO. NO increases blood flow in the gastric mucosa, promoting repair and removal of toxins [14]. NO also increases secretion of protective gastric mucus [15] and is thought to promote healing of gastric ulcers by promoting angiogenesis [16]. Alternatively, or in addition, the protective effects of NO-aspirin could be due to masking of the carboxylic acid moiety by the ester function [12, 17]. Two main subtypes of NO-aspirins have so far been developed: the nitrooxy ester (organic nitrate) derivatives and the furoxan derivatives. The first NO-aspirin hybrid drugs to be synthesised, the NicOx® compounds, NCX4016 (3-(nitroxymethyl)phenyl 2-(acetoxy)benzoate; Fig. 1a) and the related, NCX4215 [18] are both nitrooxy ester derivatives of aspirin, often referred to as "nitroaspirins". More recently, another series of NO-aspirin hybrid drugs, the furoxan derivatives, which utilise a furoxan NO-donor moiety have been developed [12, 19]. These drugs link an NO-donating moiety (furoxan group) by ester linkage to the aspirin molecule (Fig. 1b). Furoxan hybrids of aspirin with NO donor moieties have shown some benefit in avoiding acute gastric injury [12], and to be effective antiplatelet agents [19]. As the compounds have been demonstrated to inhibit COX, they have the potential to retain an aspirin-like anti-inflammatory action. Through their NO release [19], these drugs gain further potential to be anti-inflammatory through the multiple actions of NO [for review see; [20]].

Figure 1
figure 1

Structural formulae: a. The structural formula of NO-aspirin, NCX4016. b. The general structural formula of a furoxan-aspirin hybrid drug. c. The structural formulae of furoxans (B8 and B7), their NO-free equivalents, the furazans (B16 and B15) and, for comparison, aspirin.

This manuscript explores the impact of two of these novel hybrid compounds and of the furazan analogues, devoid of NO-release capacity (Fig. 1c), on release of the pro-inflammatory cytokine, TNFα from human monocyte-derived macrophages and monocytes. Cytokines are polypeptide or glycoprotein factors that act in an autocrine and/or paracrine fashion to signal in a variety of biological processes. They are generally considered to be either pro-inflammatory (e.g. TNFα and interleukin (IL)-8) or anti-inflammatory [e.g. TGFβ and IL-10] although they can have paradoxical actions. Cytokines act in various cell types and perform diverse functions. TNFα is secreted by monocytes, macrophages and neutrophils following their stimulation by bacterial LPS. The various activities of TNFα include mediation of cell adhesion molecules [21], regulation of cell death in tumour cells [22], enhancement of neutrophil responsiveness [23], control of neutrophil adherence to the endothelium [21] and synthesis of IL-1 production by macrophages [24]. LPS is known to stimulate TNFα production via the activation of the transcription factor, NF-κB [25–27]. It is well known that anti-inflammatory glucocorticoids such as dexamethasone can inhibit release of cytokines such as TNFα from cells stimulated with LPS [28–30]. Possible mechanisms include the ability of glucocorticoids to effectively suppress pro-inflammatory transcription factors such as NF-κB [31] through stimulation of glucocorticoid receptors, which subsequently translocate to the nucleus preventing histone acetylation, a vital step in the NF-κB-induced gene transcription [31, 32]. Other mechanisms by which the glucocorticoid receptor reduces TNFα expression include decreasing mRNA stability, inducing expression of the inhibitor IκB, and altering co-factor (AP-1) activity [33]. NO and aspirin have both been previously shown to influence the release of TNFα likely through modulation of NF-κB function [34–42], thus hybrid NO-aspirins may have increased anti-inflammatory potential through the dual action of NO and aspirin moieties on this pathway. Here, we set out to determine if novel NO-releasing furoxan derivatives of aspirin possess anti-inflammatory properties in LPS-activated human monocyte-derived macrophages and monocytes through analysis of TNFα release and the NF-κB pathway. The relative contribution of the aspirin and NO moieties on these effects were also addressed through the use of NO-free furazan counterpart drugs, the NO donor, DEA/NO and aspirin compounds.

Methods

Materials

General laboratory supplies were purchased from Sigma (Poole, UK) unless otherwise stated. 2-(N,N-diethyamino)-diazenolate-2-oxide (DEA/NO; Axxora, Nottingham, U.K.) was dissolved and stored frozen in 0.01 M NaOH prior to final dilution with phosphate-buffered saline (PBS) immediately prior to use. All NO-aspirin hybrids were synthesized at the Università degli Studi di Torino, as described [12]. They were dissolved in dimethyl sulfoxide (DMSO) then diluted in PBS to give a final DMSO concentration ≤ 0.1%.

Preparation of Monocytes and Macrophages

Peripheral venous blood was drawn from the antecubital fossa of human volunteers (non-smokers; age 20–45 years). Mononuclear cells were isolated by dextran sedimentation and discontinuous Percoll gradient centrifugation as described [43]. Mononuclear cells were resuspended at a concentration of 4 × 106 cells/ml in Iscove's DMEM. Monocytes were plated out in 48-well plates at a concentration of 2 × 106 per well or 6-well plates at 12 × 106 per well. After 1 h, non-adherent cells were removed by washing wells with Hank's buffered salt solution (HBSS). Macrophages were derived from monocytes by culturing the monocytes in Iscove's DMEM (supplemented with 10% autologous serum) at 37°C for a week, with the medium being changed after 3–4 days [44].

Stimulation of Cells

For macrophages, on day 7, the medium in each well was changed and fresh medium containing 10 μM test drug or a DMSO vehicle control added to wells with or without the inflammatory stimulant, LPS (10 ng/ml). Drug concentrations were selected based on results from pilot studies and on realistic plasma concentrations, where these data are available; 10 μM was deemed a relevant plasma level for aspirin, and so was utilised in this study. In order to facilitate direct comparisons, the effects of the hybrid compounds were also investigated at 10 μM. Drugs investigated were the furoxan-aspirin hybrids (3-cyanofuroxan-4-yl)methyl 2-acetoxybenzoate (B8) and (3-carbamoylfuroxan-4-yl)methyl 2-acetoxybenzoate (B7), their respective NO-free counterparts (furazans), (4-cyanofurazan-3-yl)methyl 2-(acetoxy)benzoate (B16) and (4-carbamoylfurazan-3-yl)methyl 2-acetoxybenzoate (B15), nitroaspirin NCX4016, NO donor, DEA/NO or dexamethasone (1 μM). Cells were then incubated at 37°C for 4 h before removal of the cell supernatants, which were subsequently frozen at -70°C for future studies. The same procedure was also carried out on monocytes that had not been matured into macrophages with drug treatments and 4 h incubations taking place immediately after the final cell washing step of the isolation procedure.

Enzyme-Linked Immunosorbent Assay (ELISA)

A human TNFα ELISA kit was purchased from BD Biosciences (cat no; 550610) and performed as per kit instructions on the supernatants removed from macrophage or monocyte cultures. The absorbance of each well was read at 450 nm using a Thermo Labsystems Multiskan Ascent plate reader running Ascent software Version 2.6.

Lactate Dehydrogenase (LDH) Assay

The cytotoxic impact of the compounds was assessed by measuring release of the enzyme LDH in the supernatant using a kit purchased from Roche (cat no. 1 644 793).

Western blotting for IκBα

Using 6-well plates, mononuclear cells were prepared as above. Following washing, cells were treated with Iscove's DMEM supplemented with 10% autologous serum containing either B8 (1 μM, 10 μM, 20 μM, 100 μM), gliotoxin (0.1 μg/ml), buffer or a DMSO vehicle control (0.2%) for 30 min at 37°C. LPS (10 ng/ml) or medium was then added to appropriate wells and left to incubate for a further 45 min as described for the immunofluorescence experiment (see below). Lysates were prepared at 4°C using a protease inhibitor cocktail in TBS with 1% Nonidet P40 in order to minimise proteolysis problems. An aliquot of each lysate was used for total protein determination using a BCA protein assay kit (Pierce, Rockford, IL) and an equivalent of 24 μg of protein per well was run on a 12% SDS-PAGE gel and transferred to PVDF. Blots were blocked with 5% skimmed milk in TBS/0.1% Tween-20 before probing with rabbit IκB-α (AbCam, cat no. 32518-100) [45] diluted 1:2500, incubated overnight at 4°C. Subsequently, the blots were washed and incubated with goat anti-rabbit HRP (DakoCytomation, cat no. P0448), also diluted 1:2500, and developed using standard ECL (GE Healthcare).

Immunofluorescence for NF-κB p65 Subunit

Immunofluorescence for the p65 subunit was used to visualise the translocation of NF-κB. Mononuclear cells were isolated and resuspended as described above. 4 × 106 (1 ml) cells were placed on a glass coverslip within a 6-well plate and left in an incubator (37°C; 1 h) to adhere. Non-adherent cells were then removed by washing wells with HBSS. Following washing, medium was changed to Iscove's DMEM (supplemented with 10% autologous serum) containing either gliotoxin (0.1 μg/ml), B8 or B16 (20 μM) or no drug (DMSO 0.1% control). Cells were left to incubate (37°C) for 30 min. Drug concentration and incubation times were chosen from pilot studies that demonstrated them to give optimum results in this system. LPS (10 ng/ml) or vehicle (Iscove's DMEM supplemented with 10% autologous serum) was then added on top of the coverslip and left to incubate for a further 45 min. The cell-covered coverslips were then washed 3 times with PBS and 1 ml of 3% paraformaldehyde (in deionised water; dH2O) was added and the cells left to fix (RT) for 20 min. Coverslips were then washed a further 3 times with PBS and subsequently incubated for 10 min at RT with 1 ml of 50 mM glycine to quench any aldehyde groups. Following another 3 PBS washes, 1 ml of blocking solution (10% sheep serum in 0.2% fish skin gelatin (Sigma) was added to the coverslips and they were left overnight at 4°C. The following morning, after washing the cells, 100 μl of primary antibody (NF-κB p65 mouse anti-human, BD Biosciences cat no. 610868 [46]; 1:50 dilution in blocking solution) was added to the cells. 1 h later, following three PBS washes, 100 μl of a 1:250 dilution (in blocking solution) of secondary antibody (Alexa Fluor® 488 goat anti-mouse IgG, Invitrogen cat no. A-11001) was added and left to incubate for 1 h at RT in the dark. Finally, three PBS washes, followed by three dH2O washes were carried out to prevent crystal formation. Coverslips were then mounted onto slides using Moviol (Calbiochem, Merck, Nottingham, UK) and the slides were then stored in the dark at 4°C. Images from the immunofluorescence slides were captured with a camera connected to a Zeiss Axiovert S100 microscope using Improvision Openlab 3.1.5 software. Images were captured at a magnification of ×100.

Statistical analyses

Statistical analysis was by 1-way analysis of variance (ANOVA) with Dunnett's post-test where applicable and was performed using GraphPad Prism version 4 (GraphPad Software, San Diego, USA). ** is used to represent a P value < 0.01, * denotes a P value < 0.05 and P values greater than 0.05 were deemed not significant. Where expressed, data are in the form mean ± standard error of the mean (S.E.M.).

Results

Effect on NO-furoxans on TNFα Release

B8 had a significant inhibitory effect on TNFα release in human monocyte-derived macrophages treated with LPS (36 ± 10% of LPS control, P <0.01; n = 5–10 separate donors, Fig. 2). The effect was equivalent in magnitude to that of dexamethasone, but was not shared by DEA/NO, B7, the furazans, aspirin or NCX4016. In monocytes, B8, and to a lesser extent, its NO-free equivalent, B16, significantly inhibited TNFα release [to 28 ± 5% (P < 0.01), and 49 ± 9% (P < 0.05) of control respectively, n = 4–6). Basal TNFα levels were 0.9 ± 0.2 pg/ml for macrophages and 1.6 ± 0.4 pg/ml for monocytes. After LPS-treatment these rose to 5.5 ± 0.5 and 9.6 ± 0.3 ng/ml respectively.

Figure 2
figure 2

Effect of potential anti-inflammatory agents on LPS (10 ng/ml)-induced TNFα release in human (a) monocyte-derived macrophages and (b) monocytes after 4 h treatment with 10 μM of either DEA/NO, B8, B16, B7, B15, NCX4016 or aspirin or 1 μM of dexamethasone (Dex). For macrophages n = 5–10 and for monocytes n = 4–6 separate donors. ** = p < 0.01 * = p < 0.05 determined by One-Way ANOVA followed by Dunnett's test.

Effect on NO-furoxans on LDH Release

None of the treatments studied caused significant cell death (Fig. 3) compared with untreated macrophages and monocytes. Levels of LDH released following the treatments were comparable with that from untreated control samples (~0.5 × 105 cells/treatment).

Figure 3
figure 3

Bar graphs show LDH measurement in (a) monocyte-derived macrophages and (b) monocyte supernatants after treatment with 10 μM of either DEA/NO, B8, B16, B7, B15, NCX4016 or aspirin or 1 μM of dexamethasone (Dex) and stimulated with LPS (10 ng/ml). For macrophages n = 5–15 and for monocytes n = 6 separate donors. One-way ANOVA revealed that there were no significant differences between groups in either cell type.

Effect on NO-furoxans on NF-κB activation

In order to investigate the potential molecular mechanism of action of the inhibitory effect of B8 on macrophage TNF-α release we investigated the effect of the compound on LPS-stimulated NF-κB activation. For this we assessed the effect of B8 on the loss of the cytoplasmic inhibitory subunit of NF-κB, IκBα (Fig. 4a and 4b shows two typical western blots of cytoplasmic IκBα). In both blots LPS causes a dramatic loss of cytoplasmic IκBα, an effect that was completely inhibited by the NF-κB inhibitor gliotoxin. Similarly, B8 at 100 μM also blocked LPS-induced loss of cytoplasmic IκBα (Fig. 4a and 4b) whereas 1 and 10 μM B8 (Fig. 4b) did not affect LPS loss of IκB. The concentration of B8 that appears to be on the threshold of inhibition appears to be around 20 μM (Fig. 4a). In order to confirm that B8 inhibits NF-κB activation more directly, we used immunofluorescence to investigate its effect on LPS stimulation of NF-κB p65 subunit translocation from the cytoplasm to the nucleus. NF-κB p65 immunofluorescence revealed that the subunit location varied according to the drug treatment. Control cells displayed uniform staining throughout the cytoplasm, but following stimulus with LPS, strong staining was observed in the nucleus and much less in the cytoplasm (Fig. 5a, b). Incubation with gliotoxin before the LPS stimulus inhibited the nuclear translocation of p65 as demonstrated by the presence of cytoplasmic staining (Fig. 5c). Pre-incubation with the NO-aspirin B8 gave a dramatic shift in the staining compared to the LPS control. Location of the NF-κB p65 subunit was now revealed to be cytoplasmic (Fig. 5d). Cells treated with the NO-free equivalent of B8, B16, displayed cytoplasmic staining but with a greater amount of nuclear staining than B8 treated cells (Fig. 5e).

Figure 4
figure 4

Western blots showing the effects of varying concentrations of B8 on IκB α. Mononuclear cells were prepared as per methods, plated on 6-well plates, allowed to adhere for 1 h then treated with varying concentrations of B8 (1 μM, 10 μM, 20 μM, 100 μM), gliotoxin (0.1 μg/ml), buffer or a DMSO vehicle control (0.2%) for 30 min at 37°C. After this interval LPS (10 ng/ml) or buffer was added to appropriate wells and left to incubate for a further 45 min. Lysates were prepared, total protein determined and 24 μg of protein per well was run on a 12% SDS-PAGE gel, transferred to PVDF. Blots were blocked before probing with rabbit IκB-α diluted 1:2500, incubated overnight at 4°C. Subsequently, the blots were washed and incubated with goat anti-rabbit HRP, also diluted 1:2500, then developed using standard ECL. Blots are representative of at least 6 similar experiments.

Figure 5
figure 5

Representative immunofluorescence images. Image a shows a control treated monocyte. Image b shows a monocyte following a 45 min LPS stimulus. Image c shows a monocyte treated with gliotoxin (0.1 μg/ml, 30 min) and stimulated with LPS (10 ng/ml, 45 min). Image d shows a monocyte treated with B8 (20 μM, 30 min) and stimulated with LPS (10 ng/ml, 45 min). Image e shows a monocyte treated with B16 (20 μM, 30 min) and stimulated with LPS (10 ng/ml, 45 min). C = cytoplasm. N = nucleus. Scale bar represents 100 μm. Similar images were obtained on at least 3 separate experiments.

Discussion

Impact of NO-furoxans on TNFα-Release by Macrophages and Monocytes

The NO-aspirin, B8, had a significant inhibitory effect on TNF-α release from human monocyte-derived macrophages treated with LPS. In monocytes B8, and to a lesser extent, its NO-free equivalent, B16 again caused significant inhibition of TNF-α release. In monocyte-derived macrophages, the lack of effects of B16 and aspirin suggest that the inhibitory effect of B8 on TNFα release is NO-mediated. However, as this effect is not mimicked by the NO-donor, DEA/NO, it is apparently a specific property of B8 that is possibly related to amount, duration or site of NO release. The possibility of diminished release of TNFα by B8-treated cells being simply due to a cytotoxic effect of the compound was not supported by results from the LDH assay. None of the treatments significantly affected cell death when compared with untreated cells.

In monocytes, TNFα release was inhibited by the NO-aspirin, B8, but also by its NO-free furazan counterpart, B16. Aspirin showed no significant difference from the LPS control. Similar to the macrophage results, DEA/NO did not cause significant inhibition. Again, B8 could be acting via an NO-mediated mechanism specific to the amount, duration or site of NO release. However, the interesting observation that NO-free, B16 also causes a significant inhibition suggests a possible further mechanism. As previously demonstrated, the acetyl group of these compounds is lost in plasma [12], leaving salicylic acid, through which, inhibition of cytokine release has been reported [47–49]. It may therefore be possible that under these experimental conditions, a salicylic acid-mediated mechanism is responsible for the inhibition of TNFα release observed with B8- and B16-treated cells. It is probable that the result obtained with B8 is achieved through combination of the effects of NO (as illustrated by DEA/NO) and that of the NO-free B16. We have previously shown that the furoxan-aspirin B7 releases significantly less NO than B8 [19] and thus suggest that the NO release from B7 is insufficient to result in an anti-inflammatory effect similar to that of B8. Our result obtained with B16 indicates that altering the chemical structure of aspirin clearly has an impact on the ability to achieve anti-inflammatory properties in this assay. Furthermore, the alteration to the aspirin structure in B15 and B7 likely contributed to their poor performance in this assay.

In this study, aspirin did not significantly reduce TNFα release from LPS-stimulated monocytes or macrophages. This is consistent with a similar study in which aspirin failed to have an effect even at a 30-fold higher concentrations than was used in the present study [50]. A further study did report an inhibitory effect of aspirin on TNFα release from LPS-stimulated monocytes but this was at concentrations of 5–10 mM [49], which may not be representative of pharmacologically relevant plasma concentrations [51]. We show here that NCX4016 did not significantly reduce the release of TNFα. In a study carried out by others, NCX4016 did not inhibit TNFα release at the same concentration used here (10 μM), but was shown to inhibit the release of TNFα and IL-6 from LPS-stimulated macrophages at higher (100 and 300 μM) concentrations and following a 6 h incubation [50]. Despite not being affected by the soluble guanylate cyclase inhibitor ODQ, the authors suggest that the inhibitory effect of NCX4016 is NO-mediated due to the failure of aspirin to inhibit cytokine release. A further study also showed that NCX4016 (again at concentrations 10-fold higher than used in this study), inhibited the release of IL-1β and IL-18 from LPS-stimulated monocytes, via NO-mediated inhibition of the enzyme required for intracellular processing and maturation of IL-1 and IL-18 (caspase-1) activity [52]. It is likely that the differing outcomes observed between this and other studies are purely due to drug incubation time or concentrations. The concentration studied here is based on the realistic relevant plasma concentrations of aspirin [51] and so is more demonstrative of the true therapeutic potential of the drug. The results here show that at a concentration at which the furoxan compound, B8, causes a significant 72% reduction in TNFα release from monocytes and a 64% reduction from macrophages, its organic nitrate counterpart does not.

Possible explanations for the differential effects of the same treatments observed between monocytes and macrophages effect may be the differential expression and activity of receptors and signalling pathways between the two cell types. It has previously been reported that the anti-inflammatory effect of IL-4 on the release of TNFα and other cytokines, varies between LPS-stimulated monocytes and macrophages [53]. This effect is due to loss of a receptor for IL-4 during monocyte differentiation [53]. Other differences reported between monocytes and macrophages include those showing that LPS activates cytosolic PLA2 in monocytes but not in macrophages. A similar activation in monocytes, but not macrophages, is seen after LPS-stimulation in the following signalling pathways: the MAP kinase, ERK, phosphatidylinositol-3 kinase and p70S6 kinase [54, 55]. Further variations include increased expression of Ca2+-dependent protein kinase C isoforms in monocytes when compared to macrophages [56] and also that maturation into macrophages results in slower production of the cytokine, IL-1β [57]. It is, therefore, possible that changes such as these to receptor expression, signalling pathways and to the biosynthesis of cytokines, which normally occur during the maturation of monocytes with macrophages, may impact on the ability of the studied compounds to have a significant effect.

Impact of NO-furoxans on NF-κB Activation in Monocytes

The western blot and immunofluorescence experiments further suggested a possible mechanism for the B8-mediated inhibition of TNFα release. The transcription factor, NF-κB, exists as a dimer (can be either a homodimer or heterodimer). The most common form is a heterodimer composed of the p65/p50 subunits [58]. Normally, NF-κB is kept in an inactive state in the cytoplasm, bound to an inhibitory subunit named IκBα [59]. The phosphorylation of IκBα by a kinase known as IKK and the subsequent degradation of IκBα leads to the activation of the NF-κB [60]. The NF-κB dimer containing the p65 subunit; the dominant factor in the induction of the TNFα gene, then translocates from the cytoplasm to the nucleus where it activates transcription of target genes such as TNFα [25, 27, 61, 62]. To investigate the effect of the NO-aspirin on stimulus (LPS)-induced NF-κB activation, we used two independent assays; namely loss of cytoplasmic IκBα assessed by western blotting and translocation of the p65 subunit of NF-κB assessed by immunofluorescence. For consistency, LPS was used as our NF-κB-activating stimulus and epipolythiodioxoperazine (gliotoxin), a known immunosuppressive agent that inhibits NF-κB activation by preventing IκBα degradation [43, 63], was used as the positive control. LPS caused a dramatic loss of cytoplasmic IκBα and translocation of cytoplasmic p65 to the nucleus, effects that were inhibited by NO-aspirin B8 and the positive control gliotoxin. Thus, the inhibition of NF-κB activation provides a plausible explanation for the B8-induced reduction in TNFα release as observed in the ELISA studies. It has been shown that NO inhibits LPS-induced IκB-phosphorylation and inhibits the activation of NF-κB [35], further supporting our paradigm of NO-mediated inhibition of TNFα release by B8. B16-treated and LPS-stimulated monocytes also displayed some evidence of cytoplasmic staining, but with more nuclear staining than its NO counterpart. This result is consistent with the ELISA data, where significant inhibition of TNFα release, albeit lesser than B8-treated cells, was observed following B16 treatment.

Therapeutic Implications

The data here show that the furoxan-aspirin compound B8 has anti-inflammatory effects in LPS-stimulated monocytes and macrophages through its reduction in NF-κB-mediated TNFα release. This action of B8 may be useful in inflammatory diseases (arthritis, Crohn's disease and asthma [64–67] where anti-TNFα therapy is, or has potential, to be of therapeutic benefit. Rheumatoid arthritis is a chronic inflammatory autoimmune disorder characterised by inflammation of the lining (synovium) of joints. Joint deterioration, together with the pain associated with synovial inflammation can lead to substantial loss of mobility. Pro-inflammatory cytokines are abundant in the joints of sufferers [65]. Anti-TNFα drugs have been recently licensed for use in arthritis in a bid to limit the contribution of this cytokine to the chronic joint inflammation [67]. Treatment of arthritis with drugs of the NSAID class is severely limited due to the gastric side-effects associated with the high doses and chronic nature of the treatment required. However it is hoped that NO-aspirins might offer a preferable alternative to therapy with conventional NSAIDs. The multifaceted actions of B8 on COX inhibition [19] the anti-TNFα effects demonstrated here, its observed resistance to gastrotoxic effects [12] and its potential for targeted intracellular release of NO [19, 68] could indicate B8 to be a promising anti-arthritic drug. A summary of the effects of the furoxan-aspirin hybrid drugs is provided in Fig. 6. A further, more speculative application for drugs such as B8 is in atherosclerosis therapy. It is now widely accepted that inflammation is a key element in atherogenesis and atherosclerotic plaque rupture leading to acute cardiovascular events such as myocardial infarction or stroke [69]. The release of TNFα by monocytes and macrophages causes various effects involved in destabilising the atherosclerotic plaque [70–74]. Such evidence may suggest that drugs with an anti-TNFα action, such as B8, may be of benefit in atherosclerosis.

Figure 6
figure 6

A schematic representation of the effects and potential uses of furoxan-aspirin hybrid drugs.

Conclusion

Taken together, these studies provide evidence that treatment with NO-aspirin B8, significantly reduced TNFα release from both LPS-stimulated monocytes and monocyte-derived macrophages. A possible mechanism for this anti-inflammatory action is through the inhibition of its transcription factor, NF-κB by NO. Such an action instils a potential for drugs of this NO-aspirin hybrid class to be utilised as anti-inflammatory agents for the treatment of a wide range of inflammatory conditions such as arthritis.

Abbreviations

ANOVA:

Analysis of variance

COX:

Cyclooxygenase

DEA/NO:

2-(N,N-diethyamino)-diazenolate-2-oxide

dH2O:

deionised water

DMSO:

Dimethyl sulfoxide

ELISA:

Enzyme-linked immunosorbent assay

HBSS:

Hanks buffered salt solution

IL:

Interleukin

LDH:

Lactate dehydrogenase

LPS:

Lipopolysaccharide

NO:

Nitric oxide

NSAID:

Nonsteroidal anti-inflammatory drugs

NF-κB:

Nuclear factor-kappa B

PBS:

Phosphate-buffered saline

RT:

Room temperature, TNFα: Tumour necrosis factor-alpha.

References

  1. Vane JR, Botting RM: The mechanism of action of aspirin. Thromb Res. 2003, 110: 255-258. 10.1016/S0049-3848(03)00379-7.

    Article  PubMed  CAS  Google Scholar 

  2. DeWitt DL, Smith WL: Primary structure of prostaglandin G/H synthase from sheep vesicular gland determined from the complementary DNA sequence. Proc Natl Acad Sci USA. 1988, 85: 1412-1416. 10.1073/pnas.85.5.1412.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  3. Roth GJ, Majerus PW: The mechanism of the effect of aspirin on human platelets. I. Acetylation of a particulate fraction protein. J Clin Invest. 1975, 56: 624-632. 10.1172/JCI108132.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  4. Roth GJ, Stanford N, Majerus PW: Acetylation of prostaglandin synthase by aspirin. Proc Natl Acad Sci USA. 1975, 72: 3073-3076. 10.1073/pnas.72.8.3073.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  5. Cameron AJ: Aspirin and gastric ulcer. Mayo Clin Proc. 1975, 50: 565-570.

    PubMed  CAS  Google Scholar 

  6. Seager JM, Hawkey CJ: ABC of the upper gastrointestinal tract: Indigestion and non-steroidal anti-inflammatory drugs. Bmj. 2001, 323: 1236-1239. 10.1136/bmj.323.7323.1236.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  7. Tramer MR, Moore RA, Reynolds DJ, McQuay HJ: Quantitative estimation of rare adverse events which follow a biological progression: a new model applied to chronic NSAID use. Pain. 2000, 85: 169-182. 10.1016/S0304-3959(99)00267-5.

    Article  PubMed  CAS  Google Scholar 

  8. Wallace JL: Nonsteroidal anti-inflammatory drugs and gastroenteropathy: the second hundred years. Gastroenterology. 1997, 112: 1000-1016. 10.1053/gast.1997.v112.pm9041264.

    Article  PubMed  CAS  Google Scholar 

  9. Robert A, Nezamis JE, Lancaster C, Hanchar AJ: Cytoprotection by prostaglandins in rats. Prevention of gastric necrosis produced by alcohol, HCl, NaOH, hypertonic NaCl, and thermal injury. Gastroenterology. 1979, 77: 433-443.

    PubMed  CAS  Google Scholar 

  10. Schoen RT, Vender RJ: Mechanisms of nonsteroidal anti-inflammatory drug-induced gastric damage. Am J Med. 1989, 86: 449-458. 10.1016/0002-9343(89)90344-6.

    Article  PubMed  CAS  Google Scholar 

  11. Whittle BJ: Mechanisms underlying gastric mucosal damage induced by indomethacin and bile-salts, and the actions of prostaglandins. Br J Pharmacol. 1977, 60: 455-460.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  12. Cena C, Lolli ML, Lazzarato L, Guaita E, Morini G, Coruzzi G, McElroy SP, Megson IL, Fruttero R, Gasco A: Antiinflammatory, gastrosparing, and antiplatelet properties of new NO-donor esters of aspirin. J Med Chem. 2003, 46: 747-754. 10.1021/jm020969t.

    Article  PubMed  CAS  Google Scholar 

  13. Fiorucci S, Del Soldato P: NO-aspirin: mechanism of action and gastrointestinal safety. Dig Liver Dis. 2003, 35 (Suppl 2): S9-19. 10.1016/S1590-8658(03)00047-1.

    Article  PubMed  CAS  Google Scholar 

  14. Wallace JL, Miller MJ: Nitric oxide in mucosal defense: a little goes a long way. Gastroenterology. 2000, 119: 512-520. 10.1053/gast.2000.9304.

    Article  PubMed  CAS  Google Scholar 

  15. Brown JF, Keates AC, Hanson PJ, Whittle BJ: Nitric oxide generators and cGMP stimulate mucus secretion by rat gastric mucosal cells. Am J Physiol. 1993, 265: G418-422.

    PubMed  CAS  Google Scholar 

  16. Ma L, Wallace JL: Endothelial nitric oxide synthase modulates gastric ulcer healing in rats. Am J Physiol Gastrointest Liver Physiol. 2000, 279: G341-346.

    PubMed  CAS  Google Scholar 

  17. Rainsford KD, Whitehouse MW: Gastric irritancy of aspirin and its congeners: anti-inflammatory activity without this side-effect. J Pharm Pharmacol. 1976, 28: 599-601.

    Article  PubMed  CAS  Google Scholar 

  18. Wallace JL, McKnight W, Del Soldato P, Baydoun AR, Cirino G: Anti-thrombotic effects of a nitric oxide-releasing, gastric-sparing aspirin derivative. J Clin Invest. 1995, 96: 2711-2718. 10.1172/JCI118338.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  19. Turnbull CM, Cena C, Fruttero R, Gasco A, Rossi AG, Megson IL: Mechanism of action of novel NO-releasing furoxan derivatives of aspirin in human platelets. Br J Pharmacol. 2006, 148: 517-526. 10.1038/sj.bjp.0706743.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  20. Wallace JL: Nitric oxide as a regulator of inflammatory processes. Mem Inst Oswaldo Cruz. 2005, 100 (Suppl 1): 5-9.

    PubMed  CAS  Google Scholar 

  21. Graves DT, Jiang Y: Chemokines, a family of chemotactic cytokines. Crit Rev Oral Biol Med. 1995, 6: 109-118.

    Article  PubMed  CAS  Google Scholar 

  22. Chang MP, Wisnieski BJ: Comparison of the intoxication pathways of tumor necrosis factor and diphtheria toxin. Infect Immun. 1990, 58: 2644-2650.

    PubMed  CAS  PubMed Central  Google Scholar 

  23. Klebanoff SJ, Vadas MA, Harlan JM, Sparks LH, Gamble JR, Agosti JM, Waltersdorph AM: Stimulation of neutrophils by tumor necrosis factor. J Immunol. 1986, 136: 4220-4225.

    PubMed  CAS  Google Scholar 

  24. Dinarello CA, Cannon JG, Wolff SM, Bernheim HA, Beutler B, Cerami A, Figari IS, Palladino MA, O'Connor JV: Tumor necrosis factor (cachectin) is an endogenous pyrogen and induces production of interleukin 1. J Exp Med. 1986, 163: 1433-1450. 10.1084/jem.163.6.1433.

    Article  PubMed  CAS  Google Scholar 

  25. Baldwin AS: The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 1996, 14: 649-683. 10.1146/annurev.immunol.14.1.649.

    Article  PubMed  CAS  Google Scholar 

  26. Kubes P, McCafferty DM: Nitric oxide and intestinal inflammation. Am J Med. 2000, 109: 150-158. 10.1016/S0002-9343(00)00480-0.

    Article  PubMed  CAS  Google Scholar 

  27. Totzke G, Essmann F, Pohlmann S, Lindenblatt C, Janicke RU, Schulze-Osthoff K: A novel member of the Ikappa B family, human Ikappa B-zeta, inhibits transactivation of p65 and its DNA binding. J Biol Chem. 2006

    Google Scholar 

  28. Oono H, Nakagawa M, Miyamoto A, Ishiguro S, Nishio A: Mechanisms underlying the enhanced elevation of IL-1beta and TNF-alpha mRNA levels following endotoxin challenge in rat alveolar macrophages cultured with low-Mg2+ medium. Magnes Res. 2002, 15: 153-160.

    PubMed  CAS  Google Scholar 

  29. Laufer S, Greim C, Bertsche T: An in-vitro screening assay for the detection of inhibitors of proinflammatory cytokine synthesis: a useful tool for the development of new antiarthritic and disease modifying drugs. Osteoarthritis Cartilage. 2002, 10: 961-967. 10.1053/joca.2002.0851.

    Article  PubMed  CAS  Google Scholar 

  30. Bleeker MW, Netea MG, Kullberg BJ, Ven-Jongekrijg Van der J, Meer Van der JW: The effects of dexamethasone and chlorpromazine on tumour necrosis factor-alpha, interleukin-1 beta, interleukin-1 receptor antagonist and interleukin-10 in human volunteers. Immunology. 1997, 91: 548-552. 10.1046/j.1365-2567.1997.00285.x.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  31. Barnes PJ: Corticosteroid effects on cell signalling. Eur Respir J. 2006, 27: 413-426. 10.1183/09031936.06.00125404.

    Article  PubMed  CAS  Google Scholar 

  32. Adcock IM, Ito K, Barnes PJ: Glucocorticoids: effects on gene transcription. Proc Am Thorac Soc. 2004, 1: 247-254. 10.1513/pats.200402-001MS.

    Article  PubMed  CAS  Google Scholar 

  33. Adcock IM, Caramori G, Ito K: New insights into the molecular mechanisms of corticosteroids actions. Curr Drug Targets. 2006, 7: 649-660. 10.2174/138945006777435344.

    Article  PubMed  CAS  Google Scholar 

  34. Hachicha M, Pouliot M, Petasis NA, Serhan CN: Lipoxin (LX)A4 and aspirin-triggered 15-epi-LXA4 inhibit tumor necrosis factor 1alpha-initiated neutrophil responses and trafficking: regulators of a cytokine-chemokine axis. J Exp Med. 1999, 189: 1923-1930. 10.1084/jem.189.12.1923.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  35. Hattori Y, Kasai K, Gross SS: NO suppresses while peroxynitrite sustains NF-kappaB: a paradigm to rationalize cytoprotective and cytotoxic actions attributed to NO. Cardiovasc Res. 2004, 63: 31-40. 10.1016/j.cardiores.2004.03.014.

    Article  PubMed  CAS  Google Scholar 

  36. Khan Q, Mehta JL: Relevance of platelet-independent effects of aspirin to its salutary effect in atherosclerosis-related events. J Atheroscler Thromb. 2005, 12: 185-190.

    Article  PubMed  CAS  Google Scholar 

  37. Kopp E, Ghosh S: Inhibition of NF-kappa B by sodium salicylate and aspirin. Science. 1994, 265: 956-959. 10.1126/science.8052854.

    Article  PubMed  CAS  Google Scholar 

  38. Matthews JR, Botting CH, Panico M, Morris HR, Hay RT: Inhibition of NF-kappaB DNA binding by nitric oxide. Nucleic Acids Res. 1996, 24: 2236-2242. 10.1093/nar/24.12.2236.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  39. Sekkai D, Aillet F, Israel N, Lepoivre M: Inhibition of NF-kappaB and HIV-1 long terminal repeat transcriptional activation by inducible nitric oxide synthase 2 activity. J Biol Chem. 1998, 273: 3895-3900. 10.1074/jbc.273.7.3895.

    Article  PubMed  CAS  Google Scholar 

  40. Tegeder I, Pfeilschifter J, Geisslinger G: Cyclooxygenase-independent actions of cyclooxygenase inhibitors. FASEB J. 2001, 15: 2057-2072. 10.1096/fj.01-0390rev.

    Article  PubMed  CAS  Google Scholar 

  41. Welters ID, Fimiani C, Bilfinger TV, Stefano GB: NF-kappaB, nitric oxide and opiate signaling. Med Hypotheses. 2000, 54: 263-268. 10.1054/mehy.1999.0032.

    Article  PubMed  CAS  Google Scholar 

  42. Yin MJ, Yamamoto Y, Gaynor RB: The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. Nature. 1998, 396: 77-80. 10.1038/23948.

    Article  PubMed  CAS  Google Scholar 

  43. Ward C, Chilvers ER, Lawson MF, Pryde JG, Fujihara S, Farrow SN, Haslett C, Rossi AG: NF-kappaB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem. 1999, 274: 4309-4318. 10.1074/jbc.274.7.4309.

    Article  PubMed  CAS  Google Scholar 

  44. Rossi AG, McCutcheon JC, Roy N, Chilvers ER, Haslett C, Dransfield I: Regulation of macrophage phagocytosis of apoptotic cells by cAMP. J Immunol. 1998, 160: 3562-3568.

    PubMed  CAS  Google Scholar 

  45. Bournazos S, Rennie J, Hart SP, Fox KA, Dransfield I: Monocyte Functional Responsiveness After PSGL-1-Mediated Platelet Adhesion Is Dependent on Platelet Activation Status. Arterioscler Thromb Vasc Biol. 2008, 28 (8): 1491-1498. 10.1161/ATVBAHA.108.167601.

    Article  PubMed  CAS  Google Scholar 

  46. Nishibe T, Parry G, Ishida A, Aziz S, Murray J, Patel Y, Rahman S, Strand K, Saito K, Saito Y, et al: Oncostatin M promotes biphasic tissue factor expression in smooth muscle cells: evidence for Erk-1/2 activation. Blood. 2001, 97: 692-699. 10.1182/blood.V97.3.692.

    Article  PubMed  CAS  Google Scholar 

  47. Feng H, Li XY, Zheng JR, Gao JW, Xu LF, Tang MY: Inhibition of the nuclear factor-kappaB signaling pathway by leflunomide or triptolide also inhibits the anthralin-induced inflammatory response but does not affect keratinocyte growth inhibition. Biol Pharm Bull. 2005, 28: 1597-1602. 10.1248/bpb.28.1597.

    Article  PubMed  CAS  Google Scholar 

  48. Mitchell JA, Saunders M, Barnes PJ, Newton R, Belvisi MG: Sodium salicylate inhibits cyclo-oxygenase-2 activity independently of transcription factor (nuclear factor kappaB) activation: role of arachidonic acid. Mol Pharmacol. 1997, 51: 907-912.

    PubMed  CAS  Google Scholar 

  49. Osnes LT, Foss KB, Joo GB, Okkenhaug C, Westvik AB, Ovstebo R, Kierulf P: Acetylsalicylic acid and sodium salicylate inhibit LPS-induced NF-kappa B/c-Rel nuclear translocation, and synthesis of tissue factor (TF) and tumor necrosis factor alfa (TNF-alpha) in human monocytes. Thromb Haemost. 1996, 76: 970-976.

    PubMed  CAS  Google Scholar 

  50. Minuz P, Degan M, Gaino S, Meneguzzi A, Zuliani V, Santonastaso CL, Soldato PD, Lechi A: NCX4016 (NO-Aspirin) has multiple inhibitory effects in LPS-stimulated human monocytes. Br J Pharmacol. 2001, 134: 905-911. 10.1038/sj.bjp.0704326.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  51. Rosenkranz B, Frolich JC: Plasma concentrations and anti-platelet effects after low dose acetylsalicylic acid. Prostaglandins Leukot Med. 1985, 19: 289-300. 10.1016/0262-1746(85)90142-8.

    Article  PubMed  CAS  Google Scholar 

  52. Fiorucci S, Santucci L, Cirino G, Mencarelli A, Familiari L, Soldato PD, Morelli A: IL-1 beta converting enzyme is a target for nitric oxide-releasing aspirin: new insights in the antiinflammatory mechanism of nitric oxide-releasing nonsteroidal antiinflammatory drugs. J Immunol. 2000, 165: 5245-5254.

    Article  PubMed  CAS  Google Scholar 

  53. Hart PH, Bonder CS, Balogh J, Dickensheets HL, Donnelly RP, Finlay-Jones JJ: Differential responses of human monocytes and macrophages to IL-4 and IL-13. J Leukoc Biol. 1999, 66: 575-578.

    PubMed  CAS  Google Scholar 

  54. Barbour SE, Wong C, Rabah D, Kapur A, Carter AD: Mature macrophage cell lines exhibit variable responses to LPS. Mol Immunol. 1998, 35: 977-987. 10.1016/S0161-5890(98)00070-4.

    Article  PubMed  CAS  Google Scholar 

  55. Rao KM: MAP kinase activation in macrophages. J Leukoc Biol. 2001, 69: 3-10.

    PubMed  CAS  Google Scholar 

  56. Monick MM, Carter AB, Gudmundsson G, Geist LJ, Hunninghake GW: Changes in PKC isoforms in human alveolar macrophages compared with blood monocytes. Am J Physiol. 1998, 275: L389-397.

    PubMed  CAS  Google Scholar 

  57. Herzyk DJ, Allen JN, Marsh CB, Wewers MD: Macrophage and monocyte IL-1 beta regulation differs at multiple sites. Messenger RNA expression, translation, and post-translational processing. J Immunol. 1992, 149: 3052-3058.

    PubMed  CAS  Google Scholar 

  58. Panwalkar A, Verstovsek S, Giles F: Nuclear factor-kappaB modulation as a therapeutic approach in hematologic malignancies. Cancer. 2004, 100: 1578-1589. 10.1002/cncr.20182.

    Article  PubMed  CAS  Google Scholar 

  59. Ghosh S, May MJ, Kopp EB: NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 1998, 16: 225-260. 10.1146/annurev.immunol.16.1.225.

    Article  PubMed  CAS  Google Scholar 

  60. Yamamoto Y, Gaynor RB: IkappaB kinases: key regulators of the NF-kappaB pathway. Trends Biochem Sci. 2004, 29: 72-79. 10.1016/j.tibs.2003.12.003.

    Article  PubMed  CAS  Google Scholar 

  61. Kaltschmidt B, Sparna T, Kaltschmidt C: Activation of NF-kappa B by reactive oxygen intermediates in the nervous system. Antioxid Redox Signal. 1999, 1: 129-144. 10.1089/ars.1999.1.2-129.

    Article  PubMed  CAS  Google Scholar 

  62. Liu H, Sidiropoulos P, Song G, Pagliari LJ, Birrer MJ, Stein B, Anrather J, Pope RM: TNF-alpha gene expression in macrophages: regulation by NF-kappa B is independent of c-Jun or C/EBP beta. J Immunol. 2000, 164: 4277-4285.

    Article  PubMed  CAS  Google Scholar 

  63. Pahl HL, Krauss B, Schulze-Osthoff K, Decker T, Traenckner EB, Vogt M, Myers C, Parks T, Warring P, Muhlbacher A, et al: The immunosuppressive fungal metabolite gliotoxin specifically inhibits transcription factor NF-kappaB. J Exp Med. 1996, 183: 1829-1840. 10.1084/jem.183.4.1829.

    Article  PubMed  CAS  Google Scholar 

  64. Carroccio A, Di Prima L, Pirrone G, Ambrosiano G, Noto D, Cefalu AB: [Anti-TNF (infliximab) treatment in Crohn disease: safety profile]. Recenti Prog Med. 2006, 97: 108-112. quiz 122.

    PubMed  Google Scholar 

  65. Christodoulou C, Choy EH: Joint inflammation and cytokine inhibition in rheumatoid arthritis. Clin Exp Med. 2006, 6: 13-19. 10.1007/s10238-006-0088-5.

    Article  PubMed  CAS  Google Scholar 

  66. Russo C, Polosa R: TNF-alpha as a promising therapeutic target in chronic asthma: a lesson from rheumatoid arthritis. Clin Sci (Lond). 2005, 109: 135-142.

    Article  CAS  Google Scholar 

  67. Siddiqui MA, Scott LJ: Spotlight on infliximab in Crohn disease and rheumatoid arthritis. BioDrugs. 2006, 20: 67-70. 10.2165/00063030-200620010-00008.

    Article  PubMed  Google Scholar 

  68. Turnbull CM, Rossi AG, Megson IL: Therapeutic effects of nitric oxide-aspirin hybrid drugs. Expert Opin Ther Targets. 2006, 10: 911-922. 10.1517/14728222.10.6.911.

    Article  PubMed  CAS  Google Scholar 

  69. Ross R: Atherosclerosis – an inflammatory disease. N Engl J Med. 1999, 340: 115-126. 10.1056/NEJM199901143400207.

    Article  PubMed  CAS  Google Scholar 

  70. Pober JS, Cotran RS: Cytokines and endothelial cell biology. Physiol Rev. 1990, 70: 427-451.

    PubMed  CAS  Google Scholar 

  71. Libby P, Sukhova G, Lee RT, Galis ZS: Cytokines regulate vascular functions related to stability of the atherosclerotic plaque. J Cardiovasc Pharmacol. 1995, 25 (Suppl 2): S9-12.

    Article  PubMed  CAS  Google Scholar 

  72. Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Unemori EN, Lark MW, Amento E, Libby P: Cytokine-stimulated human vascular smooth muscle cells synthesize a complement of enzymes required for extracellular matrix digestion. Circ Res. 1994, 75: 181-189.

    Article  PubMed  CAS  Google Scholar 

  73. Geng YJ, Wu Q, Muszynski M, Hansson GK, Libby P: Apoptosis of vascular smooth muscle cells induced by in vitro stimulation with interferon-gamma, tumor necrosis factor-alpha, and interleukin-1 beta. Arterioscler Thromb Vasc Biol. 1996, 16: 19-27.

    Article  PubMed  CAS  Google Scholar 

  74. Jovinge S, Crisby M, Thyberg J, Nilsson J: DNA fragmentation and ultrastructural changes of degenerating cells in atherosclerotic lesions and smooth muscle cells exposed to oxidized LDL in vitro. Arterioscler Thromb Vasc Biol. 1997, 17: 2225-2231.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

CMT was supported by a BHF Student Fellowship (FS/03/068).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Adriano G Rossi.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

The manuscript was written and the experiments were designed by CMT and AGR. CMT performed the TNF-α release, LDH assay and immunofluorescence experiments, PM performed the western blot experiments and TAS performed and assisted in all the experimental procedures. Hybrid drugs were synthesised and supplied by LL, CC, RF and AG. AGR and ILM supervised the experiments and oversaw manuscript construction together with SF, revising it critically for important intellectual content. All authors have given final approval of the version to be published.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Turnbull, C.M., Marcarino, P., Sheldrake, T.A. et al. A novel hybrid aspirin-NO-releasing compound inhibits TNFalpha release from LPS-activated human monocytes and macrophages. J Inflamm 5, 12 (2008). https://doi.org/10.1186/1476-9255-5-12

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1476-9255-5-12

Keywords