Title: NSAIDs: learning new tricks from old drugs Federico Díaz-González1 and Francisco Sánchez-Madrid2 1

Department of Internal Medicine, Universidad de La Laguna, Rheumatology Service,

Hospital Universitario de Canarias, Santa Cruz de Tenerife. Spain. 2National Center for Cardiovascular Research, Immunology Service, Instituto Investigaciones Sanitarias Princesa, Universidad Autónoma de Madrid. Spain

Key words: Non-steroidal anti-inflammatory drugs, L-selectin, NADPH oxidase,

Corresponding author:

Federico Díaz-González, Professor of Medicine. Department of

Internal Medicine, Universidad de La Laguna. Staff of Rheumatology, Hospital Universitario de Canárias. C/Ofra s/n, 38320, La Laguna, Santa Cruz de Tenerife. Spain e-mail: [email protected] fax: +34-922646792 List of abbreviations:

ADAM: a disintegrin and metalloproteinase domain, COX:

cyclooxigenase, ICAM-1: Intercellular adhesion molecule 1, IL-1: interleukin-1, iNOS: inducible nitric oxide synthase, LFA-1: lymphocyte function-associated antigen 1, MAPK: Mitogen-activated protein kinases, NADPH-oxidase: nicotinamide adenine dinucleotide phosphate-oxidase, NF-kappa B: nuclear factor kappa-light-chain-enhancer of activated B cells, NSAIDs: non-steroidal anti-inflammatory drugs, PC: phosphatidylcholine, PGE1: prostaglandin E1, PI3K: phosphatidylinositide 3-kinases, PPAR: peroxisome proliferatoractivated receptors, ROS: reactive oxygen species, TNF-: tumor necrosis factor-, VCAM1: Vascular-cell adhesion molecule, VLA-4: very late antigen-4 Received: 29-Oct-2014; Revised: 07-Dec-2014; Accepted: 16-Dec-2014 This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/eji.201445222.

This article is protected by copyright. All rights reserved.

1

Summary Nonsteroidal anti-inflammatory drugs (NSAIDs) comprise a heterogeneous group of pharmacological agents used for the symptomatic treatment of fever, pain and inflammation. Although the main mechanism of action of NSAIDs consists of inhibiting prostaglandin synthesis by blocking the enzyme cyclooxygenase (COX), clinical and experimental data strongly indicate the existence of additional mechanisms. Some of the COX-independent effects are related to the ability of NSAIDs to penetrate biological membranes and disrupt important molecular interactions necessary for a wide array of cellular functions, including cell adhesion. These effects, in particular those that interfere with L-selectin function in neutrophils during the inflammatory response, may contribute to the anti-inflammatory properties that NSAIDs exert in vivo. Recent contributions in this field have shown that the anti-L-selectin effect of NSAIDs is related to the NADPH-oxidase-dependent generation of superoxide anion at the plasma membrane. These findings might represent a novel approach for developing new and effective anti-inflammatory compounds with a better safety profile than the currently available NSAIDs. Introduction Nonsteroidal anti-inflammatory drugs (NSAIDs) are a heterogeneous group of therapeutic agents widely used for the symptomatic treatment of rheumatic disorders. Since the early seventies of last century, it has been widely accepted that the main mechanism of action of these compounds, which is also responsible for the main side effect of gastric mucosal damage, is inhibition of cyclooxygenase (COX), a key enzyme in prostaglandin synthesis [1]. Prostaglandins are group of hormone-like lipid compounds with a wide variety of strong physiological effects, including regulation of inflammation, pain sensitization, and platelet aggregation, among many others. However, a growing body of evidence suggests that NSAIDs have additional anti-inflammatory properties (reviewed in [2]). Some of these effects appear to be related to the ability of NSAIDs to penetrate biological membranes, as evaluated in vitro using membrane mimetic models, cell cultures and molecular dynamic

This article is protected by copyright. All rights reserved.

2

simulation systems [3, 4], where they disrupt normal signaling events and modify important processes necessary for cellular function, including cell adhesion [5, 6]. The ability of NSAIDs to interfere with either cell adhesion, for example by cleavage of epithelial cell adhesion molecule protein on tumor cells [6], or with leukocyte adhesion pathways essential for the inflammatory response, such as causing L-selectin shedding on neutrophil [5], has been described. Interestingly, this anti-adhesive effect of NSAIDs has also been shown to influence platelet adhesion, and it has been suggested that coagulation, hemostasis and thrombus formation could be modulated by these compounds independently of the release of pro-inflammatory mediators from platelets [7, 8]. In leukocytes, a group of NSAIDs, including flufenamic, meclofenamic, and mefenamic acids, diclofenac and aceclofenac has been shown to induce the downregulation of L-selectin, whereas another group including phenylbutazone and the oxicams, piroxicam and meloxicam has been shown to modulate the function of the integrin CD11b on neutrophils [5, 9, 10]. Some very recent contributions in this field have shown that the anti-L-selectin effect of NSAIDs also causes a significant anti-inflammatory response in vivo [11], and this anti-inflammatory response has been shown, in vitro in human neutrophils, be related to the NADPH-oxidase-dependent generation of superoxide anion at the plasma membrane [12]. In this work we review the “COX-centric” theory of NSAID mode of action, and then dissect the non-prostaglandin-mediated effects of NSAIDs, and how some of these, specifically those that interfere with cell adhesion, might explain the anti-inflammatory effects that such compounds exert in vivo. We also discuss how the effects of NSAIDs that do not rely on prostaglandin inhibition may represent a novel strategy for developing a new family of anti-inflammatory compounds. The therapeutic action of this new compound family would be based on decreasing cell adhesion, rather than on prostaglandin synthesis inhibition, thereby presenting a better safety profile than that of currently available NSAIDs. Recent advances in the understanding of non-prostaglandin-mediated antineoplastic [13] and neuroprotective [14, 15] effects of NSAIDs have also been shown, but fall beyond the scope of this review.

This article is protected by copyright. All rights reserved.

3

Challenging the “COX-centric” theory. In the early 1970s, it was proposed that inhibition of prostaglandin synthesis was the mechanism through which aspirin, the first member of the NSAID family, inhibited inflammation [16]. This mechanism later came to be the paradigm view of how NSAIDs exert their action. COX is a key enzyme in prostaglandin synthesis, and most known NSAIDs have been shown to inhibit COX activity. There are two highly related isoforms of COX: COX-1 and COX-2 [17]. COX-1, the “constitutive isoform”, has mainly cytoprotective effects, for instance in the production of gastric mucus and the maintenance of renal blood flow. In contrast, COX-2, the “inducible isoform”, is usually undetectable in most tissues, and its expression increases during the inflammatory response [18]. Based on their chemicals structure, there are now at least 20 different NSAIDs from six major groups available for use in humans (Table 1). All of them are absorbed completely orally, have negligible first-pass hepatic metabolism, and are tightly bound to albumin. In general, NSAIDs are weak organic acids with hydrophobic properties, which facilitate their binding to COX, since it is a membrane protein and the COX active site is located at the end of a hydrophobic channel [17, 19]. Table 1 lists the different members of the NSAID family classified by chemical structure, COX selectivity and proposed mechanism of action. The effect of NSAIDs on prostaglandin synthesis provides an explanation for most of their pharmacological actions, including their anti-pyretic, analgesic and platelet antiaggregant effects, as well as for their deleterious side effects, most notably stomach ulcer and renal insufficiency [20]. However, several lines of evidence suggest that COX blockade may not be the only or even the most important anti-inflammatory mechanism of action of this family of compounds. Evidence challenging COX inhibition as the only mechanism of the anti-inflammatory action of NSAIDs includes the observation that certain prostaglandins, such as PGE1, exert anti-inflammatory activity in vivo [21].

Secondly, non-acetylated

salicylates, a group of poor COX inhibitors [22] have a similar anti-inflammatory

This article is protected by copyright. All rights reserved.

4

effectiveness to that of efficient COX-inhibiting NSAIDs in patients with rheumatoid arthritis, pointing to a different mechanism of action [23]. Thirdly, there is a great disparity between the small doses of aspirin required to inhibit prostaglandin synthesis (a few hundred milligrams) and the higher doses required to exert an anti-inflammatory effect in vivo (several grams) [24]. Furthermore, in mice in which the gene encoding COX-2 has been disrupted, the inflammatory response is largely intact [25, 26]. Finally, COX-1-deficient mice show a weak inflammatory response to arachidonic acid, but not to tetradecanoyl phorbol acetate [27]. Surprisingly, these animals did not develop stomach ulcers spontaneously, and even developed less gastric ulceration than wild-type mice when treated with the NSAID indomethacin [27].

COX-independent anti-inflammatory actions of NSAIDs There is a well-known variability in the clinical response of patients to NSAIDs, which suggests that these agents might differ in their mechanism of action. The clinical evidence suggests that the inter-individual variation in the NSAID response cannot be explained by differences in pharmacokinetics, serum concentration or the enantiomeric state of the products [28]. Rather, these data suggest that prostaglandin-independent, NSAIDtriggered responses must play an important role in determining patients’ clinical responsiveness to these agents. During the past twenty years, the description of a number of prostaglandinindependent effects from NSAIDs has contributed to a better understanding of their antiinflammatory activity. Most of the described effects rely on the ability of NSAIDs to insert into the lipid bilayers of biological membranes. The use of a wide range of in vitro experimental techniques has provided significant information about the location of NSAIDs within membranes, and also on their effect on diverse membrane properties, such as fluidity, thickness, or phospholipid packaging, among others. These studies on the interaction between NSAIDs and cell membranes have indeed provided evidence on additional mechanisms to explain both the therapeutic action and toxicity of NSAIDs (reviewed in [3]). For example, a

This article is protected by copyright. All rights reserved.

5

recent study in AGS cells showed that NSAIDs have a strong affinity for membrane phospholipids, especially phosphatidylcholine (PC) [29]. This feature may explain the wide range of COX-independent, membrane-associated effects exerted by these compounds, including anion transport, oxidative phosphorylation and cell-cell interactions. Of special interest, in vitro data reported in several studies over two decades indicate that NSAIDs interfere with cell membrane events associated with neutrophil activation. The neutrophil events affected by NSAID insertion at the cell membrane include inhibition of cell aggregation, release of lysosomal enzymes (reviewed in [30]) and chemotaxis [31], activation of the NADPH oxidase complex and the oxidative burst in gastric mucosa cells and neutrophils [12, 32] and upregulation of integrins and conformational changes, and the shedding of L-selectin [5, 9, 33]. Other lines of investigation have postulated that the anti-inflammatory action of some NSAIDs can be related to their ability to modulate transcription factors. Some NSAIDs, including salicylates, aspirin, ibuprofen and some nitric oxide-donating aspirin derivatives, might exert some of their anti-inflammatory actions by inhibiting nuclear factor-kappa B (NF-kappa B), a key transcription factor that controls the inducible expression of many genes involved in inflammation, including those encoding pro-inflammatory cytokines such as TNF- or IL-1, iNOS and adhesion molecules such as ICAM-1 (Intercellular adhesion molecule 1) and VCAM-1 (Vascular-cell adhesion molecule 1) [34]. At high doses, aspirin and sodium salicylate are able to inhibit the activation of NF-kappa B in several cell lines by a mechanism that prevents the degradation of the NF-kappa B inhibitor, I kappa B [35]. This effect has been tested and shown to occur in different cell types, including fibroblasts, epithelial cell, or endothelial cells [36-38]. Other NSAIDs act similarly, but at concentrations comparable to those used in therapy. For example, the NSAID ibuprofen has been shown to inhibit NF-kappa B activation in human T cells, at concentrations similar to those obtained in human plasma (micromolar range) [39]. Although the NF-kappa B inhibitory effect has also been described for other NSAIDs, such as indomethacin, flurbiprofen or sulindac, it is not the

This article is protected by copyright. All rights reserved.

6

mechanism of all NSAIDs, since naproxen, a highly effective commonly used NSAID, does not affect NF-kappa B activity [40]. Several NSAIDs have been shown to significantly inhibit neutrophil chemotaxis toward CXCL8 and C5a, an effect that has been linked to the polymerization of F-actin and inhibition of the chemotactic pathway dependent on PI3K/Akt activation [41]. Other reports have claimed that NSAIDs can also regulate other signaling pathways, such as indomethacin, which has been shown to activate the MAPK pathway [42], or diclofenac and celecoxib, which have been shown to function via PPAR [43]; there is the suggestion that these actions may underlie the anti-neoplastic effects of these compounds [44]. However, the implication of these anti-tumor effects in the anti-inflammatory properties of NSAIDs has not yet been determined. All these data suggest that NSAIDs have anti-inflammatory mechanisms of action other than the inhibition of prostaglandin synthesis (Table 1). However, the role of most of these COX-independent processes in clinical inflammation remains to be clarified.

Inhibition of cell adhesion by NSAIDs Clinical experience shows that NSAIDs are more effective in acute rather than chronic inflammatory diseases, suggesting that NSAIDs interfere preferentially with the early steps of the inflammatory response. An early event essential for an effective inflammatory response is the transmigration of leukocytes across the vascular endothelium, and their accumulation in inflamed tissues. This cellular extravasation requires a succession of highly coordinated adhesion events between flowing leukocytes and endothelial cells, a process commonly known as the adhesion cascade. The adhesion cascade can be divided into four successive steps (Figure 1), each of which is essential for leukocyte extravasation, such that if one step is blocked, leukocyte emigration does not occur properly [45, 46].

This article is protected by copyright. All rights reserved.

7

A major effort in the field of inflammation is currently aimed at developing antagonists of adhesion receptors, an approach known as anti-adhesive therapy [47-49]. This strategy is based on the understanding that if any of the sequential adhesion events of the adhesion cascade is inhibited, overall inflammation will decrease, ameliorating its deleterious effects. In neutrophils, two groups of NSAIDs have been shown to have anti-adhesive properties, one interfering with the function of L-selectin, the other with CD11b/CD18 [5, 9], two adhesion molecules that play key roles in the adhesion cascade (Figure 1). Remarkably, NSAID-induced shedding of L-selectin has been demonstrated not only in vivo in both humans and mice [5, 11], but also in vitro, where the concentration of NSAIDs required to induce L-selectin shedding in human neutrophils [5, 12] are within the range (micromolars) reached in plasma by oral administration of NSAIDs in humans [50, 51].

L-selectin-based effects of NSAIDs L-selectin is constitutively expressed by most leukocytes, and is enzymatically cleaved and released after cell activation through the processing of its ectodomain by ADAM17 [52] and ADAM8 [53], members of the disintegrin and metalloproteinase domain (ADAM) family of surface metalloproteases (Figure 2). L-selectin plays a major role in the early recruitment of neutrophils to inflammatory foci by mediating their rolling on the endothelial surface [54]. Both in vitro [55] and in vivo [56] experiments have shown that soluble L-selectin is able to reduce intravascular leukocyte rolling and adhesion, and consequently decreases neutrophil extravasation presumably by competing with cell surface L-selectin to binding to endothelial ligands. A number of NSAIDs have been shown to induce rapid cleavage and shedding of Lselectin in human neutrophils in vitro [5, 10]. Analysis of the structure-function relationship of some NSAIDs has shown that the ability to downregulate L-selectin expression is dependent on the presence of diphenylamine or its related compound N-phenylanthranilic acid in the NSAID structural core [10].

This article is protected by copyright. All rights reserved.

8

The anti-L-selectin action of NSAIDs involves neither non-specific activation of neutrophils nor prostaglandin inhibition, but has been shown to require the presence of ADAM17 [10]. Shedding of L-selectin in neutrophils can be induced by oxidative attack on the pro-domain thiol group of ADAM17, which masks its functional catalytic domain [57]. A group of NSAIDs, including diclofenac and flufenamic acid, was recently shown to interfere with the ability of human neutrophils to initiate interaction with endothelial cells by triggering L-selectin-shedding through the production of reactive oxygen species (ROS), a highly oxidizing agent generated at the plasma membrane [12]. Data obtained in vitro with neutrophils from patients with chronic granulomatous disease (a hereditary defect in the NADPH oxidase complex that results in the reduction of ROS production) demonstrate that NSAIDs such as diclofenac, require NADPH oxidase-dependent generation of superoxide anion to trigger L-selectin shedding [12] (Figure 2). How NSAIDs activate NADPH oxidase remains unknown. However, given that PC has an inhibitory effect on the NADPH oxidase complex [58], it is conceivable that NSAID–PC interaction at the plasma membrane might reduce this inhibitory effect, facilitating superoxide anion production [29]. These findings are in accordance with the emerging view that ROS have regulatory functions that limit inflammation (reviewed in [59]). Several studies have investigated whether induction of L-selectin shedding by NSAIDs is sufficient to reduce inflammation in vivo. In healthy human volunteers, the basal surface expression of L-selectin on circulating neutrophils is significantly reduced by therapeutic doses of indomethacin [5]. In the zymosan air pouch model of acute inflammation, intramuscular treatment with N-phenylanthranilic acid, a diphenylaminerelated NSAID that promotes L-selectin shedding in neutrophils in vitro [10] and in vivo [11], was shown to interfere with the ability of neutrophils to accumulate at inflammatory foci [11]. At the doses used in that study, N-phenylanthranilic acid did not block COX and did not show any additive anti-inflammatory effect to treatment with Mel-14, a functional blocking monoclonal antibody against murine L-selectin [11]. These observations indicate that the anti-inflammatory effect of NSAIDs in vivo may be mediated by the induction of L-

This article is protected by copyright. All rights reserved.

9

selectin shedding. The development of new compounds specifically designed to target Lselectin might therefore be a productive strategy for controlling the pathologic inflammatory response.

NSAIDs and anti-adhesive therapy A major effort in the field of inflammation is currently directed at developing antagonists of adhesion receptors. For more than ten years the field of leukocyte cell adhesion has been considered a potential source of novel and potent targets for the treatment of inflammation. Until now, most advances in this field have been in the development of therapeutic agents that block the function of integrins. Anti-adhesive therapies targeting the major leukocyte integrins, such as LFA-1 and VLA-4, have proved relatively successful in the treatment of human inflammatory disorders, including psoriasis, Crohn’s disease and multiple sclerosis [47, 60]. However, the inhibition of selectins or their ligands has only proved beneficial in certain animal models of inflammation, such as hemorrhagic-traumatic shock in baboons [61] and ischemia/reperfusion injury in rat cells [62], with a limited clinical success in human inflammatory conditions such as experimental endotoxemia and multiple trauma (reviewed in [63]). The anti-inflammatory effect of the anti-L-selectin action of NSAIDs in vivo [11] suggests that a strategy based on inducing L-selectin shedding could offer advantages over L-selectin-ligand blockade using antibodies [64] or synthetic selectin antagonist [65]. This approach would prevent the inflammatory response through a dual action of i) reducing neutrophil L-selectin surface expression and ii) quenching endothelial Lselectin ligands by generating higher concentrations of soluble L-selectin in plasma.

Conclusions In addition to their unquestionable role in the human therapeutic arsenal, NSAIDs have made an immense contribution to our knowledge of prostaglandin physiology, platelet aggregation, cytoprotection and renal blood flow. Currently, NSAIDs continue to surprise us

This article is protected by copyright. All rights reserved.

10

with non-prostaglandin-related therapeutic effects of potential benefit in the field of neuroprotection or cancer. In the field of cell adhesion, NSAIDs have effects that not only provide a mechanistic explanation for their anti-inflammatory properties, but also provide new ideas for managing inflammation. In this review we have summarized the most important prostaglandin–independent anti-inflammatory effects of NSAIDs, emphasizing their action on L-selectin expression in neutrophils, a capability that underlies their antiinflammatory action in vivo. The induction of L-selectin shedding, either dependent on NADPH oxidase activation or by other as yet unknown mechanism, could develop into a successful anti-inflammatory treatment by preventing neutrophil rolling and consequently decreasing the intensity of the inflammatory response more effectively than L-selectin-ligand blockade alone, a tested strategy with limited clinical effects. The development of new compounds reproducing the ability of NSAIDs to induce L-selectin shedding might yield new therapeutic tools for the clinical management of inflammatory conditions.

Acknowledgements Funding for this project came from Fondo de Investigaciones Sanitarias, cofinanced by the European Regional Development Fund (FIS PI12/02499), and REUNINVES (Asociación para la Ayuda a la Investigación en Reumatología del Hospital Universitario de Canarias) to F.D-G, also from the Spanish Ministry of Science and Innovation (SAF2011-25834), Comunidad de Madrid (INDISNET-S2011/BMD-2332), Instituto Salud Carlos III (Red Cardiovascular RD 12-0042-0056), and ERC-2011-AdG 294340-GENTRIS to F-S-M. The authors thank S. Bartlett for English editing, M. Vicente-Manzanares for critical reading of the manuscript and artwork, and Mercedes Guerra for the documentation work.

Conflict of Interest Disclosure The authors declare no commercial or financial conflict of interest

This article is protected by copyright. All rights reserved.

11

References 1

Warner, T. D., Giuliano, F., Vojnovic, I., Bukasa, A., Mitchell, J. A. and Vane, J. R., Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci U S A 1999. 96: 7563-7568.

2

Gan, T. J., Diclofenac: an update on its mechanism of action and safety profile. Curr Med Res Opin 2010. 26: 1715-1731.

3

Pereira-Leite, C., Nunes, C. and Reis, S., Interaction of nonsteroidal antiinflammatory drugs with membranes: in vitro assessment and relevance for their biological actions. Prog Lipid Res 2013. 52: 571-584.

4

Khajeh, A. and Modarress, H., The influence of cholesterol on interactions and dynamics of ibuprofen in a lipid bilayer. Biochim Biophys Acta 2014. 1838: 24312438.

5

Diaz-Gonzalez, F., Gonzalez-Alvaro, I., Campanero, M. R., Mollinedo, F., del Pozo, M. A., Munoz, C., Pivel, J. P. et al., Prevention of in vitro neutrophilendothelial

attachment

through

shedding

of

L-selectin

by

nonsteroidal

antiinflammatory drugs. J Clin Invest 1995. 95: 1756-1765. 6

Liggett, J. L., Min, K. W., Smolensky, D. and Baek, S. J., A novel COXindependent mechanism of sulindac sulfide involves cleavage of epithelial cell adhesion molecule protein. Exp Cell Res 2014. 326: 1-9.

This article is protected by copyright. All rights reserved.

12

7

Dominguez-Jimenez, C., Diaz-Gonzalez, F., Gonzalez-Alvaro, I., Cesar, J. M. and Sanchez-Madrid, F., Prevention of alphaII(b)beta3 activation by non-steroidal antiinflammatory drugs. FEBS Lett 1999. 446: 318-322.

8

Zarbock, A. and Rossaint, J., L-selectin shedding by NSAIDs: old friends in new dresses. Eur J Immunol 2013. 43: 50-54.

9

Garcia-Vicuna, R., Diaz-Gonzalez, F., Gonzalez-Alvaro, I., del Pozo, M. A., Mollinedo, F., Cabanas, C., Gonzalez-Amaro, R. et al., Prevention of cytokineinduced changes in leukocyte adhesion receptors by nonsteroidal antiinflammatory drugs from the oxicam family. Arthritis Rheum 1997. 40: 143-153.

10

Gomez-Gaviro, M. V., Gonzalez-Alvaro, I., Dominguez-Jimenez, C., Peschon, J., Black, R. A., Sanchez-Madrid, F. and Diaz-Gonzalez, F., Structure-function relationship and role of tumor necrosis factor-alpha-converting enzyme in the downregulation of L-selectin by non-steroidal anti-inflammatory drugs. J Biol Chem 2002. 277: 38212-38221.

11

Herrera-Garcia, A., Dominguez-Luis, M., Arce-Franco, M., Lopez-Fernandez, J., Feria, M., Barreiro, O., Sanchez-Madrid, F. et al., In vivo modulation of the inflammatory response by nonsteroidal antiinflammatory drug-related compounds that trigger L-selectin shedding. Eur J Immunol 2013. 43: 55-64.

12

Dominguez-Luis, M., Herrera-Garcia, A., Arce-Franco, M., Armas-Gonzalez, E., Rodriguez-Pardo, M., Lorenzo-Diaz, F., Feria, M. et al., Superoxide anion mediates the L-selectin down-regulation induced by non-steroidal anti-inflammatory drugs in human neutrophils. Biochem Pharmacol 2013. 85: 245-256.

This article is protected by copyright. All rights reserved.

13

13

Dovizio, M., Tacconelli, S., Sostres, C., Ricciotti, E. and Patrignani, P., Mechanistic and pharmacological issues of aspirin as an anticancer agent. Pharmaceuticals 2012. 5: 1346-1371.

14

Ajmone-Cat, M. A., Bernardo, A., Greco, A. and Minghetti, L., Non-steroidal anti-inflammatory drugs and brain inflammation: Effects on microglial functions. Pharmaceuticals 2010. 3: 1949-1964.

15

Weggen, S., Rogers, M. and Eriksen, J., NSAIDs: small molecules for prevention of Alzheimer's disease or precursors for future drug development? Trends Pharmacol Sci 2007. 28: 536-543.

16

Vane, J. R., Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol 1971. 231: 232-235.

17

Simmons, D. L., Botting, R. M. and Hla, T., Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacol Rev 2004. 56: 387-437.

18

Dubois, R. N., Abramson, S. B., Crofford, L., Gupta, R. A., Simon, L. S., Van De Putte, L. B. and Lipsky, P. E., Cyclooxygenase in biology and disease. Faseb J 1998. 12: 1063-1073.

19

Smith, W. L., Urade, Y. and Jakobsson, P. J., Enzymes of the cyclooxygenase pathways of prostanoid biosynthesis. Chem Rev 2011. 111: 5821-5865.

20

Ballou, L. R. and Wang, B. W. E., Nonsteroidal Anti-inflammatory Drugs. In Firestein, G. S., Budd, R. C., Harris, E. D., MacInnes, I. B., Ruddy, S. and Sergent, J. S. (Eds.) Kelley´s Textbook of Rheumatology, 8th Edn. Saunders Elsevier, Philadelphia 2009, pp 833-862.

This article is protected by copyright. All rights reserved.

14

21

Fantone, J. C., Marasco, W. A., Elgas, L. J. and Ward, P. A., Anti-inflammatory effects of prostaglandin E1: in vivo modulation of the formyl peptide chemotactic receptor on the rat neutrophil. J Immunol 1983. 130: 1495-1497.

22

Mitchell, J. A., Akarasereenont, P., Thiemermann, C., Flower, R. J. and Vane, J. R., Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase. Proc Natl Acad Sci U S A 1993. 90: 11693-11697.

23

Bombardier, C., Peloso, P. M. and Goldsmith, C. H., Salsalate, a nonacetylated salicylate, is as efficacious as diclofenac in patients with rheumatoid arthritis. Salsalate-Diclofenac Study Group. J Rheumatol 1995. 22: 617-624.

24

Abramson, S. B. and Weissmann, G., The mechanisms of action of nonsteroidal antiinflammatory drugs. Arthritis Rheum 1989. 32: 1-9.

25

Dinchuk, J. E., Car, B. D., Focht, R. J., Johnston, J. J., Jaffee, B. D., Covington, M. B., Contel, N. R. et al., Renal abnormalities and an altered inflammatory response in mice lacking cyclooxygenase II. Nature 1995. 378: 406-409.

26

Morham, S. G., Langenbach, R., Loftin, C. D., Tiano, H. F., Vouloumanos, N., Jennette, J. C., Mahler, J. F. et al., Prostaglandin synthase 2 gene disruption causes severe renal pathology in the mouse. Cell 1995. 83: 473-482.

27

Langenbach, R., Morham, S. G., Tiano, H. F., Loftin, C. D., Ghanayem, B. I., Chulada, P. C., Mahler, J. F. et al., Prostaglandin synthase 1 gene disruption in mice reduces arachidonic acid-induced inflammation and indomethacin-induced gastric ulceration. Cell 1995. 83: 483-492.

This article is protected by copyright. All rights reserved.

15

28

Walker, J. S., Sheather-Reid, R. B., Carmody, J. J., Vial, J. H. and Day, R. O., Nonsteroidal antiinflammatory drugs in rheumatoid arthritis and osteoarthritis: support for the concept of "responders" and "nonresponders". Arthritis Rheum 1997. 40: 1944-1954.

29

Lichtenberger, L. M., Zhou, Y., Jayaraman, V., Doyen, J. R., O'Neil, R. G., Dial, E. J., Volk, D. E. et al., Insight into NSAID-induced membrane alterations, pathogenesis and therapeutics: characterization of interaction of NSAIDs with phosphatidylcholine. Biochim Biophys Acta 2012. 1821: 994-1002.

30

Altman, R. D., Neutrophil activation: an alternative to prostaglandin inhibition as the mechanism of action for NSAIDs. Semin Arthritis Rheum 1990. 19: 1-5.

31

Bertolotto, M., Pende, A., Ottonello, L., Montecucco, F. and Dallegri, F., Treatment with oxaprozin reduces neutrophil migration in response to C5a and CXCL8 via the inhibition of the PI3K/Akt pathway. Eur J Clin Invest 2013. 43: 29.

32

Tanaka, J., Yuda, Y. and Yamakawa, T., Mechanism of superoxide generation system in indomethacin-induced gastric mucosal injury in rats. Biol Pharm Bull 2001. 24: 155-158.

33

Pillinger, M. H., Capodici, C., Rosenthal, P., Kheterpal, N., Hanft, S., Philips, M. R. and Weissmann, G., Modes of action of aspirin-like drugs: salicylates inhibit erk activation and integrin-dependent neutrophil adhesion. Proc Natl Acad Sci U S A 1998. 95: 14540-14545.

34

Barnes, P. J. and Karin, M., Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997. 336: 1066-1071.

This article is protected by copyright. All rights reserved.

16

35

Yin, M. J., Yamamoto, Y. and Gaynor, R. B., The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. Nature 1998. 396: 77-80.

36

Sakurada, S., Kato, T. and Okamoto, T., Induction of cytokines and ICAM-1 by proinflammatory cytokines in primary rheumatoid synovial fibroblasts and inhibition by N-acetyl-L-cysteine and aspirin. Int Immunol 1996. 8: 1483-1493.

37

Yoo, C. G., Lee, S., Lee, C. T., Kim, Y. W., Han, S. K. and Shim, Y. S., Effect of acetylsalicylic acid on endogenous I kappa B kinase activity in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2001. 280: L3-9.

38

Weber, C., Erl, W., Pietsch, A. and Weber, P. C., Aspirin inhibits nuclear factorkappa B mobilization and monocyte adhesion in stimulated human endothelial cells. Circulation 1995. 91: 1914-1917.

39

Scheuren, N., Bang, H., Munster, T., Brune, K. and Pahl, A., Modulation of transcription factor NF-kappaB by enantiomers of the nonsteroidal drug ibuprofen. Br J Pharmacol 1998. 123: 645-652.

40

Kazmi, S. M., Plante, R. K., Visconti, V., Taylor, G. R., Zhou, L. and Lau, C. Y., Suppression of NF kappa B activation and NF kappa B-dependent gene expression by tepoxalin, a dual inhibitor of cyclooxygenase and 5-lipoxygenase. J Cell Biochem 1995. 57: 299-310.

41

Bertolotto, M., Contini, P., Ottonello, L., Pende, A., Dallegri, F. and Montecucco, F., Neutrophil migration towards C5a and CXCL8 is prevented by non-steroidal antiinflammatory drugs via inhibition of different pathways. Br J Pharmacol 2014. 171: 3376-3393.

This article is protected by copyright. All rights reserved.

17

42

Ou, Y. C., Yang, C. R., Cheng, C. L., Raung, S. L., Hung, Y. Y. and Chen, C. J., Indomethacin induces apoptosis in 786-O renal cell carcinoma cells by activating mitogen-activated protein kinases and AKT. Eur J Pharmacol 2007. 563: 49-60.

43

Vaish, V., Tanwar, L. and Sanyal, S. N., The role of NF-kappaB and PPARgamma in experimentally induced colorectal cancer and chemoprevention by cyclooxygenase2 inhibitors. Tumour Biol 2010. 31: 427-436.

44

Kashfi, K. and Rigas, B., Non-COX-2 targets and cancer: expanding the molecular target repertoire of chemoprevention. Biochem Pharmacol 2005. 70: 969-986.

45

Barreiro, O., Martin, P., Gonzalez-Amaro, R. and Sanchez-Madrid, F., Molecular cues guiding inflammatory responses. Cardiovasc Res 2010. 86: 174-182.

46

Ley, K., Laudanna, C., Cybulsky, M. I. and Nourshargh, S., Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 2007. 7: 678-689.

47

Ghosh, S. and Panaccione, R., Anti-adhesion molecule therapy for inflammatory bowel disease. Therap Adv Gastroenterol 2010. 3: 239-258.

48

Griffith, J. W. and Luster, A. D., Targeting cells in motion: migrating toward improved therapies. Eur J Immunol 2013. 43: 1430-1435.

49

Sunasee, R., Adokoh, C. K., Darkwa, J. and Narain, R., Therapeutic potential of carbohydrate-based polymeric and nanoparticle systems. Expert Opin Drug Deliv 2014. 11: 867-884.

This article is protected by copyright. All rights reserved.

18

50

Basmenji, S., Valizadeh, H. and Zakeri-Milani, P., Comparative in vitro dissolution and in vivo bioequivalence of two diclofenac enteric coated formulations. Arzneimittelforschung 2011. 61: 566-570.

51

Kokki, H., Kumpulainen, E., Laisalmi, M., Savolainen, J., Rautio, J. and Lehtonen, M., Diclofenac readily penetrates the cerebrospinal fluid in children. Br J Clin Pharmacol 2008. 65: 879-884.

52

Peschon, J. J., Slack, J. L., Reddy, P., Stocking, K. L., Sunnarborg, S. W., Lee, D. C., Russell, W. E. et al., An essential role for ectodomain shedding in mammalian development. Science 1998. 282: 1281-1284.

53

Gomez-Gaviro, M., Dominguez-Luis, M., Canchado, J., Calafat, J., Janssen, H., Lara-Pezzi, E., Fourie, A. et al., Expression and regulation of the metalloproteinase ADAM-8 during human neutrophil pathophysiological activation and its catalytic activity on L-selectin shedding. J Immunol 2007. 178: 8053-8063.

54

Smalley, D. M. and Ley, K., L-selectin: mechanisms and physiological significance of ectodomain cleavage. J Cell Mol Med 2005. 9: 255-266.

55

Schleiffenbaum, B., Spertini, O. and Tedder, T. F., Soluble L-selectin is present in human plasma at high levels and retains functional activity. J Cell Biol 1992. 119: 229-238.

56

Ferri, L. E., Pascual, J., Seely, A. J., Chaudhury, P. and Christou, N. V., Soluble L-selectin attenuates tumor necrosis factor-alpha-mediated leukocyte adherence and vascular permeability: a protective role for elevated soluble L-selectin in sepsis. Crit Care Med 2002. 30: 1842-1847.

This article is protected by copyright. All rights reserved.

19

57

Zhang, Z., Oliver, P., Lancaster, J. R., Jr., Schwarzenberger, P. O., Joshi, M. S., Cork, J. and Kolls, J. K., Reactive oxygen species mediate tumor necrosis factor alpha-converting, enzyme-dependent ectodomain shedding induced by phorbol myristate acetate. Faseb J 2001. 15: 303-305.

58

Gorzalczany, Y., Sigal, N., Itan, M., Lotan, O. and Pick, E., Targeting of Rac1 to the phagocyte membrane is sufficient for the induction of NADPH oxidase assembly. J Biol Chem 2000. 275: 40073-40081.

59

Hultqvist, M., Olsson, L. M., Gelderman, K. A. and Holmdahl, R., The protective role of ROS in autoimmune disease. Trends Immunol 2009. 30: 201-208.

60

Hyun, Y. M., Lefort, C. T. and Kim, M., Leukocyte integrins and their ligand interactions. Immunol Res 2009. 45: 195-208.

61

Schlag, G., Redl, H. R., Till, G. O., Davies, J., Martin, U. and Dumont, L., Anti-Lselectin antibody treatment of hemorrhagic-traumatic shock in baboons. Crit Care Med 1999. 27: 1900-1907.

62

Takada, M., Nadeau, K. C., Shaw, G. D., Marquette, K. A. and Tilney, N. L., The cytokine-adhesion molecule cascade in ischemia/reperfusion injury of the rat kidney. Inhibition by a soluble P-selectin ligand. J Clin Invest 1997. 99: 2682-2690.

63

Mackay, C. R., Moving targets: cell migration inhibitors as new anti-inflammatory therapies. Nat Immunol 2008. 9: 988-998.

64

Seekamp, A., van Griensven, M., Dhondt, E., Diefenbeck, M., Demeyer, I., Vundelinckx, G., Haas, N. et al., The effect of anti-L-selectin (aselizumab) in

This article is protected by copyright. All rights reserved.

20

multiple traumatized patients--results of a phase II clinical trial. Crit Care Med 2004. 32: 2021-2028. 65

Mayr, F. B., Firbas, C., Leitner, J. M., Spiel, A. O., Reiter, R. A., Beyer, D., Meyer, M. et al., Effects of the pan-selectin antagonist bimosiamose (TBC1269) in experimental human endotoxemia. Shock 2008. 29: 475-482.

66

Alon, R., Kassner, P. D., Carr, M. W., Finger, E. B., Hemler, M. E. and Springer, T. A., The integrin VLA-4 supports tethering and rolling in flow on VCAM-1. J Cell Biol 1995. 128: 1243-1253.

67

Amin, A. R., Vyas, P., Attur, M., Leszczynska-Piziak, J., Patel, I. R., Weissmann, G. and Abramson, S. B., The mode of action of aspirin-like drugs: effect on inducible nitric oxide synthase. Proc Natl Acad Sci U S A 1995. 92: 7926-7930.

68

Xu, X. M., Sansores-Garcia, L., Chen, X. M., Matijevic-Aleksic, N., Du, M. and Wu, K. K., Suppression of inducible cyclooxygenase 2 gene transcription by aspirin and sodium salicylate. Proc Natl Acad Sci U S A 1999. 96: 5292-5297.

69

Gomez-Gaviro, M. V., Dominguez-Jimenez, C., Carretero, J. M., Sabando, P., Gonzalez-Alvaro, I., Sanchez-Madrid, F. and Diaz-Gonzalez, F., Down-regulation of L-selectin expression in neutrophils by nonsteroidal anti-inflammatory drugs: role of intracellular ATP concentration. Blood 2000. 96: 3592-3600.

70

Tegeder, I., Niederberger, E., Israr, E., Guhring, H., Brune, K., Euchenhofer, C., Grosch, S. et al., Inhibition of NF-kappaB and AP-1 activation by R- and Sflurbiprofen. Faseb J 2001. 15: 595-597.

This article is protected by copyright. All rights reserved.

21

71

Gonzalez-Alvaro, I., Munoz, C., Garcia-Vicuna, R., Sabando, P., Cabanas, C., Sanchez-Madrid, F. and Diaz-Gonzalez, F., Interference of nonsteroidal antiinflammatory drugs with very late activation antigen 4/vascular cells adhesion molecule 1-mediated lymphocyte-endothelial cell adhesion. Arthritis Rheum 1998. 41: 1677-1688.

72

Yamamoto, Y., Yin, M. J., Lin, K. M. and Gaynor, R. B., Sulindac inhibits activation of the NF-kappaB pathway. J Biol Chem 1999. 274: 27307-27314.

73

Shen, Y., Yang, T., Wang, J., Xu, Q., Li, R., Pan, W., Li, G. et al., Indomethacin enhances the cytotoxicity of recombinant human lymphotoxin alpha on tumor cells by suppressing NFkappaB signaling. Cancer Biol Ther 2007. 6: 1428-1433.

74

Gonzalez-Alvaro, I., Carmona, L., Diaz-Gonzalez, F., Gonzalez-Amaro, R., Mollinedo, F., Sanchez-Madrid, F., Laffon, A. et al., Aceclofenac, a new nonsteroidal antiinflammatory drug, decreases the expression and function of some adhesion molecules on human neutrophils. J Rheumatol 1996. 23: 723-729.

This article is protected by copyright. All rights reserved.

22

Figure 1. The adhesion cascade. Members of three major cell-surface adhesion receptor families are implicated in leukocyte extravasation: selectins (represented by L-selectin, blue, left), integrins (represented by LFA-1, grey, left), and the immunoglobulin superfamily (not shown). In response to tissue injury, proinflammatory mediators such as IL-1 and TNF- are released that induce a rapid change in the adhesive properties of the endothelial cells (EC) lining the blood vessels near the damaged tissue. Flowing leukocytes (Lk) are immediately captured and begin to roll over the endothelial surface (right). This initial leukocyte– endothelium interaction is mainly mediated by selectins. L-selectin (CD62L) (constitutively expressed by most leukocytes), E-selectin (CD62E) and P-selectin (CD62P) (both expressed by activated endothelial cells) specifically interact with carbohydrate moieties linked to mucin-like molecules expressed by activated endothelial cells and leukocytes (green). The selectin–mucin interaction is responsible for the rolling of leukocytes along the endothelium. During rolling, leukocytes are activated by locally produced chemokines bound to the endothelium, which cause both the shedding of L-selectin, and the activation of the integrin adhesion receptors CD11a/CD18 and CD11b/CD18, as well as the increased expression of CD11b/CD18. The activated leukocyte integrins (open conformation) interact with their endothelial counter-receptor intercellular adhesion molecule (ICAM)-1 (orange), resulting in the firm adhesion of leukocytes to the vessel surface. Finally, leukocytes migrate across the endothelium into the tissues, usually by squeezing between endothelial cells. Several endothelial junctional proteins participate in this final step, including CD31, CD99 and JAMs (junctional adhesion molecules). All of these interactions are required for neutrophil transmigration, but in the case of lymphocytes the rolling and firm adhesion steps are mainly mediated by interaction between the integrin adhesion receptor very late activation antigen (VLA)-4 and its endothelial counter receptor vascular cell adhesion molecule (VCAM)-1 [66]. NSAIDs have been found to interfere with molecules essential for the rolling or firm adhesion steps of the adhesion cascade.

This article is protected by copyright. All rights reserved.

23

This article is protected by copyright. All rights reserved.

24

Figure 2. NSAID activity on L-selectin shedding from neutrophils: a proposed mechanism. Superoxide anion production by the NADPH oxidase complex is proposed to drive NSAID-induced L-selecting shedding from neutrophils. NSAIDs insert into the lipid bilayer of the cell membrane where they interact with phospholipids, such as phosphatidylcholine (PC, green). PC is capable of interfering with the activation of the NADPH oxidase complex (orange), and it is likely that NSAID–PC interaction at the plasma membrane ablates the inhibitory effects of PC, causing activation of NADPH oxidase and consequently the release of superoxide anion to the extracellular milieu. Superoxide generates an oxidative attack on the pro-domain thiol group of ADAM-17 (grey), which masks its catalytic domain. Upon removal of this mask, ADAM17 is activated and able to process the extracellular domain of L-selectin (blue), causing its release from the cell membrane.

This article is protected by copyright. All rights reserved.

25

Table 1. NSAID families Drug

Mechanism of action

Reference

 Inhibition of PG synthesis  L-selectin sheeding  Inhibition of NFKappa B  Inhibition of iNOS  Inhibition of COX-2 gene transcription  L-selectin sheeding  Inhibition of NFKappa B  Inhibition of iNOS

[16]

 Inhibition of prostaglandin synthesis  Inhibition of NFKappa B  Inhibition of neutrophil PI3K/Aktdependent chemotactic pathways  L-selectin sheeding  Inhibition of NFKappa B  L-selectin sheeding  Inhibition of neutrophil PI3K/Aktdependent chemotactic pathways

[1]

 Inhibition of prostaglandin synthesis  L-selectin sheeding

[1]

Non-selective NSAID agents Salicylate Aspirin (acetylated)

Salicylates (non-acetylated)

[5, 10] [35] [67] [68] [69] [35] [67]

Diflunisal Salsalate Propionic acids

Ibuprofen Naproxen

Ketoprofen Flurbiprofen

Oxaprozin

Acetic acids

Diclofenac

This article is protected by copyright. All rights reserved.

[39] [41]

[5] [70] [10] [41]

[5]

26

 Inhibition of VLA-4

[71]

activation Etodolac Tolmetin Sulindac

 Inhibition of NF-

[72]

Kappa B  Cleavage of epithelial

[6]

cell adhesion molecule Ketorolac Indomethacin

 Inhibition of NF-

[73]

Kappa B  Induction of L-selectin

[5]

shedding  Inhibition of VLA-4

[71]

activation Aceclofenac

 Induction of L-selectin

[69, 74]

shedding  Inhibition of VLA-4

[71]

activation  Inhibition of

Oxicams (enolic acid)

[1]

prostaglandin synthesis Piroxicam

 Inhibition of 2

[9]

integrin activation Meloxicam

 Inhibition of 2

[9]

integrin activation

Fenamates (anthanilic acid)

 Inhibition of

[1]

prostaglandin

This article is protected by copyright. All rights reserved.

27

synthesis Meclofenami c acid Mefenamic acid

Non-acidic Nabumetone (naphthylbutanone

 Induction of L-selectin

[10]

shedding  Induction of L-selectin

[10]

shedding  Inhibition of

[1]

prostaglandin

)

synthesis by its active metabolite: 6methoxy-2-naphthyl acetic acid  Inhibition of

Selective COX-2 inhibitors

[1]

prostaglandin synthesis Celecoxib Eterocoxib

This article is protected by copyright. All rights reserved.

28

NSAIDs: learning new tricks from old drugs.

Nonsteroidal anti-inflammatory drugs (NSAIDs) comprise a heterogeneous group of pharmacological agents used for the symptomatic treatment of fever, pa...
797KB Sizes 5 Downloads 5 Views