26
Review Article NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen Species Rajeshwary Ghosh, 1 Azra Alajbegovic, 1 and Aldrin V. Gomes 1,2 1 Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA 2 Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA Correspondence should be addressed to Aldrin V. Gomes; [email protected] Received 22 December 2014; Revised 3 March 2015; Accepted 3 March 2015 Academic Editor: Mark J. Crabtree Copyright © 2015 Rajeshwary Ghosh et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a variety of conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. e beneficial effects of NSAIDs in reducing or relieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented. e undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack and stroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have been shown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increases in ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathways is apoptosis which causes cell death when significantly activated. is review discusses the relationship between NSAIDs and cardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD. 1. Introduction Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most widely used over-the-counter drugs as well as the most prescribed class of drugs for a variety of conditions including pains, rheumatoid arthritis, osteoarthritis, musculoskeletal disorders, and other comorbid conditions [1]. Millions of people suffer from pain resulting in the prolonged use of NSAIDs being common. Besides reducing or relieving pain NSAIDs have been shown to be useful as anticancer agents in various kinds of cancers [24]. However, NSAIDs also have undesirable side effects including ulcers [5], bleeding [6], kid- ney failure [7, 8], and increased risk of heart attack and stroke [8, 9]. One of the mechanisms which has been associated with the adverse effects of NSAIDs is the generation of oxidative stress. e present review focuses on NSAIDs-induced ROS generation leading to cardiovascular diseases (CVD). 2. Types of NSAIDs NSAIDs may be classified according to their mechanism of action. Nonselective NSAIDs like ibuprofen and naproxen, which comprise one class, inhibit both cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) enzymes. A second class of NSAIDs (celecoxib and rofecoxib) targets only the COX-2 pathway and is termed as COX-2 selective inhibitors (also known as coxibs). COX selectivity is one of the determining factors that is considered when administrating NSAIDs to a patient. Administration of nonselective NSAIDs has been associated with side effects like peptic ulcer disease and gastrointestinal bleeding [10]. COX-2 selective NSAIDs have been shown to exhibit gastroprotective effects unlike the nonselective NSAIDs and are thus useful in patients with painful gastrointestinal conditions [1012]. Another class of semiselective NSAIDs (indomethacin, meloxicam, and diclofenac) have a higher affinity for COX-2 but tend to inhibit the COX-1 pathway also [13]. However, irrespective of their mechanism of action, prolonged exposure to any class of NSAIDs has been shown to have potential adverse effects on cardiovascular events in patients with or without preexisting cardiovascular conditions, depending on the duration and dosage of these drugs [14, 15] (Table 1). Patients with preexisting cardiovascular conditions such as coronary Hindawi Publishing Corporation Oxidative Medicine and Cellular Longevity Volume 2015, Article ID 536962, 25 pages http://dx.doi.org/10.1155/2015/536962

Review Article NSAIDs and Cardiovascular Diseases: Role of ... › journals › omcl › 2015 › 536962.pdfReview Article NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Review ArticleNSAIDs and Cardiovascular Diseases:Role of Reactive Oxygen Species

    Rajeshwary Ghosh,1 Azra Alajbegovic,1 and Aldrin V. Gomes1,2

    1Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA2Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA

    Correspondence should be addressed to Aldrin V. Gomes; [email protected]

    Received 22 December 2014; Revised 3 March 2015; Accepted 3 March 2015

    Academic Editor: Mark J. Crabtree

    Copyright © 2015 Rajeshwary Ghosh et al. This is an open access article distributed under the Creative Commons AttributionLicense, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properlycited.

    Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a varietyof conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. The beneficial effects of NSAIDs in reducing orrelieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented.The undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack andstroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have beenshown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increasesin ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathwaysis apoptosis which causes cell death when significantly activated. This review discusses the relationship between NSAIDs andcardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD.

    1. Introduction

    Nonsteroidal anti-inflammatory drugs (NSAIDs) are themost widely used over-the-counter drugs as well as the mostprescribed class of drugs for a variety of conditions includingpains, rheumatoid arthritis, osteoarthritis, musculoskeletaldisorders, and other comorbid conditions [1]. Millions ofpeople suffer from pain resulting in the prolonged use ofNSAIDs being common. Besides reducing or relieving painNSAIDs have been shown to be useful as anticancer agents invarious kinds of cancers [2–4]. However, NSAIDs also haveundesirable side effects including ulcers [5], bleeding [6], kid-ney failure [7, 8], and increased risk of heart attack and stroke[8, 9]. One of themechanismswhich has been associatedwiththe adverse effects of NSAIDs is the generation of oxidativestress. The present review focuses on NSAIDs-induced ROSgeneration leading to cardiovascular diseases (CVD).

    2. Types of NSAIDs

    NSAIDs may be classified according to their mechanism ofaction. Nonselective NSAIDs like ibuprofen and naproxen,

    which comprise one class, inhibit both cyclooxygenase-1(COX-1) and cyclooxygenase-2 (COX-2) enzymes. A secondclass of NSAIDs (celecoxib and rofecoxib) targets only theCOX-2 pathway and is termed as COX-2 selective inhibitors(also known as coxibs). COX selectivity is one of thedetermining factors that is considered when administratingNSAIDs to a patient. Administration of nonselective NSAIDshas been associated with side effects like peptic ulcer diseaseand gastrointestinal bleeding [10]. COX-2 selective NSAIDshave been shown to exhibit gastroprotective effects unlikethe nonselective NSAIDs and are thus useful in patientswith painful gastrointestinal conditions [10–12]. Anotherclass of semiselective NSAIDs (indomethacin, meloxicam,and diclofenac) have a higher affinity for COX-2 but tend toinhibit the COX-1 pathway also [13]. However, irrespectiveof their mechanism of action, prolonged exposure to anyclass of NSAIDs has been shown to have potential adverseeffects on cardiovascular events in patients with or withoutpreexisting cardiovascular conditions, depending on theduration and dosage of these drugs [14, 15] (Table 1). Patientswith preexisting cardiovascular conditions such as coronary

    Hindawi Publishing CorporationOxidative Medicine and Cellular LongevityVolume 2015, Article ID 536962, 25 pageshttp://dx.doi.org/10.1155/2015/536962

  • 2 Oxidative Medicine and Cellular Longevity

    Table1:Listof

    NSA

    IDsw

    iththeirrecom

    mendeddo

    sesa

    ndlevelsin

    thec

    irculation.

    Genericname

    Chem

    icalname

    Recommendeddo

    sages

    (may

    vary

    depend

    ingon

    thea

    geandcond

    ition

    )

    Levelsin

    thec

    irculation(𝜇M

    and𝜇g/mL)

    (dosed

    ependent)

    Traden

    ame

    Diclofenac

    {2-[(2,6-D

    ichlorop

    henyl)a

    mino]ph

    enyl

    acetic

    acid

    25mg–150m

    g/day

    * 5𝜇M

    (1.5𝜇g/mL)

    and7𝜇

    M(2.0𝜇g/mL)

    [for5

    0mgand75

    mgdo

    ses,respectiv

    ely]

    Zorvolex,C

    ataflam

    ,Cam

    bia,Vo

    ltaren,

    Voltarengel,Arthrotec

    (com

    binedwith

    miso

    prostol),

    Flector,Zipsor,

    Penn

    said

    Indo

    methacin

    [1-(4-C

    hlorob

    enzoyl)-5-metho

    xy-2-

    methyl-1H-in

    dol-3

    -yl]a

    cetic

    acid

    25mg–200m

    g/day

    * 3𝜇M–6𝜇M

    (1to

    2𝜇g/mL)

    Tivorbex,Ind

    ocin,Ind

    ocin

    SR,

    Indo

    -Lem

    mon

    ,Ind

    omethagan

    Ketoprofen

    2-(3-Benzoylph

    enyl)p

    ropano

    icacid

    25mg–300m

    g/day

    **2𝜇

    Mto

    22𝜇M

    (0.43to

    5.62𝜇g/mL)

    [50m

    gdo

    se]

    Oruvail,Nexcede

    Ibup

    rofen

    2-(4-Isobu

    tylphenyl)prop

    anoica

    cid

    100m

    g–800m

    g/day

    * 97𝜇

    M–291𝜇M

    (20–

    60𝜇g/mL)

    [400

    mg

    dose]

    Advil,Motrin

    andNurofen,V

    icop

    rofen

    (com

    binedwith

    hydrocod

    one),D

    uexis

    (com

    binedwith

    famotidine)

    Naproxen

    (2S)-2-(6-Metho

    xy-2-naphthyl)prop

    anoic

    acid

    250m

    g–1000

    mg/day

    (bothoralandIV

    )

    * 377𝜇M

    (95𝜇

    g/mL)

    [500

    mgdo

    seon

    ceevery12hor

    1000

    mg

    dose

    for5

    days]

    Naprosyn,

    EC-N

    aprosyn,

    Naprapac

    (cop

    ackagedwith

    lansop

    razole),Aleve,

    Accord,A

    naprox

    DS,An

    aproxAntalgin,

    Apranax,Trexim

    et(com

    binedwith

    sumatrip

    tansuccinate)andVimovo

    (com

    binedwith

    esom

    eprazolemagnesiu

    m)

    Sulin

    dac

    {(1Z)-5-Fluo

    ro-2-m

    ethyl-1-[4-

    (methylsu

    lfinyl)b

    enzylid

    ene]-1H-in

    den-3-

    yl}acetic

    acid

    200m

    g–40

    0mg/day

    **1.4𝜇M–6𝜇M

    (0.5–2.0𝜇g/mL)[con

    stant

    fora

    lldo

    ses]

    Clinoril

    Ketorolac

    5-Be

    nzoyl-2

    ,3-dihydro-1H-pyrrolizine-1-

    carboxylic

    acid

    10mg–40

    mg/day

    **39𝜇M

    (10𝜇

    g/mL)

    Torado

    l,Sprix

    Piroxicam

    4-Hydroxy-2-m

    ethyl-N

    -(2-pyrid

    inyl)-2H

    -1,2

    -benzothiazine-3-carbo

    xamide1,1-

    dioxide

    0.2m

    g–20

    mg/day

    * 5𝜇M

    to6𝜇

    M(1.5to

    2𝜇g/mL)

    [single

    20mgdo

    se]9𝜇M

    to24𝜇M

    (3–8𝜇g/mL)

    [for2

    0mgrepeated

    doses]

    Feldene

  • Oxidative Medicine and Cellular Longevity 3

    Table1:Con

    tinued.

    Genericname

    Chem

    icalname

    Recommendeddo

    sages

    (may

    vary

    depend

    ingon

    thea

    geandcond

    ition

    )

    Levelsin

    thec

    irculation(𝜇M

    and𝜇g/mL)

    (dosed

    ependent)

    Traden

    ame

    Nim

    esulide

    N-(4-Nitro-2-

    phenoxyphenyl)m

    ethanesulfo

    namide

    100m

    gtwiced

    aily

    **10𝜇M

    to20𝜇M

    (2.86to

    4.58)m

    g/L

    [100

    mg]

    [175]

    Sulid

    e,Nim

    alox,M

    esulid,X

    ilox,Nise

    ,Nexen,

    Nidolon

    Diflun

    isal

    2,4-D

    ifluo

    ro-4-hydroxy-3-

    biph

    enylcarboxylic

    acid

    250m

    g–1000

    mg

    **164𝜇

    M(41𝜇

    g/mL)

    [250

    mgdo

    ses]

    348𝜇

    M(87𝜇

    g/mL)

    [500

    mgdo

    ses]and

    496𝜇

    M(124𝜇g/mL)

    [single100

    0mgd

    ose]

    Dolob

    id

    Flurbiprofen

    2-(2-Fluoro-4-biph

    enylyl)propano

    icacid

    50mg–300m

    g/day

    **60𝜇M

    (14mg/L)

    [0.65m

    g/kg

    offlu

    rbiprofenIV

    ][176]

    Ansaid

    Mefenam

    icacid

    2-[(2,3-

    Dim

    ethylphenyl)a

    mino]benzoicacid

    250m

    gfollo

    wed

    by500m

    gevery6ho

    urs

    * 83𝜇

    M(20𝜇

    g/mL)

    [250

    mgsin

    gled

    oses]

    Ponstel

    Tolm

    etin

    [1-Methyl-5

    -(4-methylbenzoyl)-1H

    -pyrrol-

    2-yl]acetic

    acid

    5mg–1800

    mg/day

    **156𝜇

    M(40𝜇

    g/mL)

    [400

    mgoraldo

    se]

    Tolectin,Tolectin

    DS,To

    lectin

    600

    Thetablelistson

    lythoseNSA

    IDsthath

    avebeen

    mentio

    nedin

    Table3alon

    gwith

    theirrecom

    mendeddo

    sesa

    ndrespectiv

    econcentrations

    inthecirculation.

    Mosto

    fthe

    inform

    ationregardingthedo

    sagesa

    ndlevelsin

    serum

    orplasmaof

    theNSA

    IDsh

    asbeen

    takenfro

    mtheFD

    Aapproved

    site,http://www.drugs.c

    om.N

    imesulideistheon

    lyNSA

    IDin

    thelistthatisn

    otavailablein

    theUSA

    .Referencesfor

    theplasma

    concentrations

    offlu

    rbiprofenandnimesulidea

    regivenin

    thetable.

    * Serum

    concentrations,*

    * Plasm

    acon

    centratio

    ns.

  • 4 Oxidative Medicine and Cellular Longevity

    Naproxen, ibuprofen, ketoprofen

    (cytosolic/secreted) (induced by thrombin, injury/inflammation,

    hormones like epinephrine, angiotensin II)

    Cyclooxygenase pathway

    COX-1(constitutive)

    COX-2(inducible)

    Valdecoxib, celecoxib, rofecoxib(Coxibs)

    NSAIDS

    Prostaglandin G2

    Prostacyclin(endothelium)

    NSAIDS

    PGD

    synt

    hase

    Thro

    mbo

    xane

    synt

    hase

    PGI s

    ynth

    ase

    PGF synthase

    PGE synthase

    Potent inhibitorof platelet

    aggregation,vasodilator

    Potent activator and stimulator of

    platelet aggregation, vasoconstrictor

    (smooth muscle)

    Arachidonic acid

    Phospholipase A2

    Protein degradation Protein synthesis

    Leukotriene

    COXIBS

    Atherosclerosis, acute myocardial

    infarction, thrombosis

    Platelet aggregation

    Lipoxygenase pathway

    Homeostasis disruption

    COX-1, COX-2

    COOHCH3

    Cytokines IL-1,IFN-𝛾, growth

    factors

    PGG2

    Prostaglandin H2PGH2

    PGF2

    Homeostasis maintained by PGF2 and PGE2

    (brain)neurophysiological

    functions

    PGD2TXA2

    TXB2

    PGI2

    (platelets)

    TXA2 PGI2

    PGE2

    Membrane phospholipid

    Figure 1: Inhibition of cyclooxygenase pathway by NSAIDs. Coxibs as well as nonselective NSAIDs inhibit the formation of the metabolitesof the cyclooxygenase pathway thereby disrupting the homeostasis maintained by these metabolites. Coxibs cause an imbalance betweenthe levels of thromboxane and prostacyclin being more favorable towards thromboxane and decreasing prostacyclin levels leading to theaggregation of platelets and causing thrombosis.

    artery disease, hypertension, and history of stroke are at thegreatest risk of cardiovascular events after taking NSAIDs[14, 15]. Patients who have recently had cardiovascular bypasssurgery are advised not to take NSAIDs due to a high riskof heart attacks [16, 17]. The increased selectivity for COX-2 has also been reported to increase the risk of various CVD[18, 19].Meta-analyses of several trials have shown that coxibs

    are associated with a high risk of atherothrombotic vascularevents [20].

    3. Mechanism of Action of NSAIDs

    NSAIDs exert their pain relieving effect mainly by inhibit-ing the cyclooxygenase pathway (Figure 1). This pathway

  • Oxidative Medicine and Cellular Longevity 5

    is responsible for the conversion of arachidonic acid toprostaglandins and thromboxanes [21]. Although COX isofficially known as prostaglandin-endoperoxide synthase(PTGS) the abbreviation “COX” is commonly used forcyclooxygenase-1 and cyclooxygenase-2 in medicine. Ingenetics, the abbreviation PTGS is officially used for thecyclooxygenase family of genes and proteins to preventambiguity with the cytochrome c oxidase family of genes andproteins which are also abbreviated COX. Arachidonic acidis the main precursor to the formation of various eicosanoidsin the cyclooxygenase pathway. Of all the metabolites formedin the arachidonic acid metabolism, thromboxane A2 in theplatelets is the major product which along with prostacy-clin (prostaglandin I2) maintains vascular homeostasis [22](Figure 1). Both these eicosanoids (thromboxane A2 andprostaglandin I2) have opposing effects. While thromboxaneis well known for its role in vasoconstriction and aggregationof platelets, prostacyclin is important for platelet aggregationinhibition and vasodilation.

    The COX enzyme is present as two isoforms, each withdistinct functions: COX-1 is constitutively expressed in thestomach, kidneys, intestinal mucosa, and other tissues [23].It protects the mucosal lining of the stomach and playsan important role in vasoconstriction and platelet aggre-gation [23]. On the other hand the inducible COX-2 isupregulated during times of inflammation where it causesvasodilation [24]. COX-1 andCOX-2 are similar inmolecularweights, 70 and 72 kDa, respectively, and show65%homologywith near-identical catalytic sites (based upon informationfrom UniProtKB/Swiss-Prot database). The critical differ-ence between the isoenzymes, which permits the selectiveinhibition of each isoform, is the substitution of isoleucine523 in COX-1 with valine in COX-2 [25]. The presenceof valine, which is a smaller amino acid than isoleucine,allows drugs entrance to a hydrophobic side-pocket onlyaccessible in COX-2. Expression of both isoforms, COX-1and COX-2, may be upregulated and downregulated undervarious pathological conditions [26]. It is likely that theclassification of the COX enzymes into two isoforms wasan oversimplification [26] as COX-2 may be constitutivelyexpressed in the brain [27], kidney [28], and testes [29]. In factimmunohistochemical studies have revealed the constitutiveexpression of COX-2 mRNA in the lung, thyroid gland,spleen, and adipose tissue, which was greater than COX-1 inthese tissues, and in the liver both isoforms were expressedequally [29]. Therefore the idea that COX-2 can only beexpressed under inducible conditions is unlikely since recentevidence suggests their occurrence in various human tissuesunder normal conditions.

    Coxibs disrupt the balance between the levels of throm-boxane A2 and prostaglandin I2 leading to atherosclerosis,thrombosis, and other cardiovascular complications. Coxibs,through their selective inhibition of COX-2, inhibit endothe-lial cell synthesis of prostacyclin [30]. In the ApoE−/−mice (model for atherosclerosis), deletion of the prostacyclinreceptor increased atherogenesis with no such effect observedin thromboxane receptor deleted mice [30]. Apart from itsrole in the inhibition of cyclooxygenase pathway, NSAIDshave been shown to cause cell death by the inhibition of

    the Akt signaling pathway [31], downregulation of the NF-𝜅Bpathway [32], downregulation of the Bcl pathway [33], upreg-ulation of the nonsteroidal activated gene-1 [34], and alteringthe p53 pathway [35], all of which have been suggested tobe involved in apoptosis [36]. Apoptosis (programmed celldeath) induced by NSAIDs has been suggested to be due tooxidative stress caused by increased generation of reactiveoxygen species (ROS) [37].

    A series of mechanisms are involved wherein NSAIDsexert their cardiotoxic effects and cause various cardiacconditions. Various non-NSAID drugs like doxorubicin,azidothymidine, and cisplatin have been shown to induceoxidative stress as a consequence of elevated ROS levels [38].Doxorubicin, for example, induced cardiotoxicity throughDNA damage and apoptosis in cardiac cells as a result ofoxidative stress which were reduced by the antioxidant effectof statin [37]. It is possible that the oxidative stress induced byNSAIDs, which is known to cause apoptosis and cell death, issignificantly involved in causing cardiovascular dysfunction(Figure 2).

    4. Incidences of CVD Induced by NSAIDs

    Various clinical trials have been made during the past fewyears regarding the safety and effectiveness of NSAIDs inCVD (Table 2). Most trials focused on the CVD outcome ofNSAID use in patients with a previous history of cardiovas-cular disease. Few trials were carried out on patients withno history of CVD. An important finding is that not onlynonselective NSAIDs lead to the development of hyperten-sion in both normotensive and hypertensive individuals [39],but their use interferes with the antihypertensivemedicationsexcept for the calcium channel blockers [40].The risk of atrialfibrillation, heart failure, myocardial infarction, and othercardiovascular conditions also increased in patients with ahistory of these pathological conditions (Table 2).

    The CVD related outcomes in patients enrolled in theREACH (REduction of Atherothrombosis for ContinuedHealth) registry showed that in patients with establishedstable atherothrombosis, use of NSAIDs increased the inci-dences of myocardial infarction and cerebrovascular con-ditions [41]. Additionally, this paper reported that the useof NSAIDs with other antiplatelet drugs (except aspirin)increased the rate of cardiovascular events like cardiovasculardeath, myocardial infarction, and stroke [41]. Although a fewstudies suggest that COX selectivity does not seem to be adetermining factor for myocardial infarction [13, 42], severalstudies suggest that coxibs elevate the rate of incidences ofCVD compared to nonselective NSAIDs [18–20]. Severalclinical trials have been completed and are still ongoing, butsome inconsistencies in the results exist regarding the effectof different types of NSAIDs on cardiovascular outcomes(Table 2). One meta-analysis report found that ∼75% studiesinvestigating the cardiovascular risk in new NSAID usersreported an increase in the occurrence of cardiovasculardiseases within the first month of NSAID use [43]. Overall,the different trials showed that several NSAIDs increasedthe risk of CVD both at low and high doses [41, 43–45].Targeted inhibition of COX-2, even for a short term, was

  • 6 Oxidative Medicine and Cellular Longevity

    Table2:Summaryof

    results

    from

    clinicaltria

    lson

    thea

    dverse

    effectsof

    NSA

    IDso

    ncardiovascular

    diseases.

    NSA

    IDClinicaltrialn

    ame/locatio

    nDurationof

    study

    Num

    bero

    fpatientsselected

    Effect

    References

    Cele

    coxib,naproxen

    sodium

    ,and

    placebo

    Alzh

    eimer’sDise

    ase

    Anti-Infl

    ammatory

    Preventio

    nTrial

    (ADAPT

    )/USA

    4years

    2,528patie

    ntsw

    ithafam

    ilyhisto

    ryof

    Alzh

    eimer’s

    Dise

    ase.

    CVD/CBV

    death,MI,str

    oke,CH

    F,or

    TIAin

    thec

    elecoxib-,naproxen-,and

    placebo-tre

    ated

    grou

    pswe

    re5.54%,

    8.25%,and

    5.68%,respectively

    .Death

    ratesinthec

    aseo

    fpatientstaking

    NSA

    IDsw

    ereh

    igherb

    utno

    tstatistic

    ally

    significant

    [44]

    Cele

    coxib,rofecoxib,valdecoxib,

    diclo

    fenac,naproxen,ibu

    profen,

    diflu

    nisal,etod

    olac,fenop

    rofen,

    flurbiprofen,

    indo

    methacin,

    ketoprofen,

    ketoralac,meclofenamate,mefenam

    icacid,m

    eloxicam,n

    abum

    eton

    e,oxaprozin,

    piroxicam,sulindac,andtolm

    etin

    Non

    e/USA

    5years

    74,838

    userso

    fNSA

    IDsa

    nd23,53

    5userso

    fother

    drugs.

    Thea

    djustedrateratio

    forR

    ofecoxib

    was

    1.16forM

    Iand

    1.15forstro

    kewhich

    was

    theh

    ighestam

    ongallother

    NSA

    IDs

    inclu

    ding

    otherc

    oxibs.Naproxen

    mod

    estly

    redu

    cedther

    ater

    atio

    (RRfor

    MI0

    .067

    andRR

    forstro

    ke0.083)

    ofcardiovascular

    events

    [177]

    Cele

    coxib,ibup

    rofen

    Non

    e/Ch

    ieti,

    Chieti,

    Italy

    3mon

    ths

    24patie

    ntsu

    ndergoing

    aspirin

    treatmentfor

    cardioprotectio

    n

    Ibup

    rofeninterfe

    redwith

    theinh

    ibition

    ofplateletCO

    X-1n

    ecessary

    for

    cardioprotectio

    nby

    aspirin

    [178]

    Diclofenac,ibup

    rofen,

    indo

    methacin,

    ketoprofen,n

    aproxen,

    mefenam

    icacid,

    piroxicam,tenoxicam

    ,tolfenamicacid,

    aceclofenac,tia

    profenicacid,m

    efenam

    icacid,etodo

    lac,nabu

    meton

    e,nimesulide,

    andmelo

    xicam,rofecoxib,cele

    coxib,

    valdecoxib,and

    etoricoxib

    Non

    e/Finland

    3years

    33,309

    patie

    ntsw

    ithfirst

    MI,138,949controls

    TheirresultssuggestthatC

    OX-

    selectivity

    dono

    tdeterminethe

    adversee

    ffectof

    CVDby

    NSA

    IDs,atleastcon

    cerningMI.

    NoNSA

    IDisMI-protectiv

    e

    [13]

    Etoricoxib

    anddiclo

    fenac

    Multin

    ationalE

    toric

    oxib

    andDiclofenacA

    rthritis

    Long

    -term

    (MED

    AL)

    Stud

    yProgram/M

    ultin

    ational

    18mon

    ths

    34,701

    patie

    nts(24,913

    with

    osteoarthritisa

    nd9,7

    87with

    rheumatoidarthritis)

    Thrombo

    ticcardiovascular

    events

    occurred

    in320patie

    ntsinthee

    toric

    oxib

    grou

    pand323patie

    ntsinthed

    iclofenac

    grou

    pwith

    eventrates

    of1.2

    4and1.3

    0per

    100patie

    nt-yearsandah

    azardratio

    of0.95

    fore

    toric

    oxibcomparedto

    diclo

    fenac.Lo

    ngterm

    usageo

    feith

    erof

    theseN

    SAID

    shad

    similare

    ffectso

    nthe

    rateso

    fthrom

    botic

    cardiovascular

    events

    [179]

    Etoricoxib

    anddiclo

    fenac

    Etoricoxibversus

    DiclofenacS

    odium

    Gastro

    intestinal

    Tolerabilityand

    Effectiv

    eness(ED

    GE)

    trial/U

    SA

    9.3mon

    thsfor

    etoricoxib

    and8.9

    mon

    thsfor

    diclo

    fenac

    7,111patie

    nts

    etoricoxib

    (𝑛=3,593)o

    rdiclo

    fenacs

    odium

    (𝑛=3,518)

    Ther

    ateo

    fthrom

    botic

    CVeventswas

    1.30and1.2

    4in

    userso

    fetoric

    oxib

    (90m

    g)anddiclo

    fenac(150m

    g),

    respectiv

    ely,w

    ithin

    28days

    [180]

  • Oxidative Medicine and Cellular Longevity 7

    Table2:Con

    tinued.

    NSA

    IDClinicaltrialn

    ame/locatio

    nDurationof

    study

    Num

    bero

    fpatientsselected

    Effect

    References

    Ibup

    rofen,

    naproxen

    orlumira

    coxib

    TheTh

    erapeutic

    Arthritis

    Research

    and

    Gastro

    intestinalEv

    entT

    rial

    (TARG

    ET)/International

    Extend

    edrepo

    rt18,32

    5patie

    ntsw

    ithosteoarthritis

    Ibup

    rofenincreasedris

    kof

    thrombo

    sisandCH

    Fcomparedto

    lumira

    coxib

    (2.14

    %versus

    0.25%)a

    mon

    gaspirin

    users.Naproxenconferslow

    erris

    krelativetolumira

    coxibam

    ongno

    naspirin

    users

    [181]

    Diclofenac,ibup

    rofen,

    andnaproxen

    Non

    e/USA

    7years

    ND

    Prolon

    gedexpo

    sure

    todiclo

    fenac

    increasesthe

    riskof

    AMI(relativer

    atio

    =1.9

    –2.0)u

    nlikeibu

    profen

    ornaproxen

    [182]

    Etoricoxib

    anddiclo

    fenac

    EDGEII/U

    SA19.3mon

    thsfor

    etoricoxib

    and19.1

    mon

    thsfor

    diclo

    fenac

    4,086patie

    ntsw

    ithrheumatoidarthritis

    etoricoxib𝑛=2,032;

    diclo

    fenac𝑛=2,054

    Ther

    ateo

    foccurrenceo

    fAMIw

    ashigh

    erin

    diclo

    fenac(0.68)treated

    patie

    ntsthan

    inetoricoxib

    users(0.43).Also

    anoverall

    increase

    incardiace

    ventsw

    asob

    served

    indiclo

    fenactreated

    grou

    p(1.14

    )versus

    thee

    toric

    oxibgrou

    p(0.83)

    [183]

    Cele

    coxib(at4

    00mgQD,200

    mgtwo

    times

    aday

    (BID

    ),or

    400m

    gBID)

    Non

    e/USA

    3years

    7,950

    patie

    nts(with

    arthritisandother

    cond

    ition

    s)

    Theh

    azardratio

    forthe

    CVD,M

    I,HF,or

    thrombo

    embo

    liceventw

    aslowestfor

    the

    400mgon

    cead

    ay(1.1),intermediatefor

    the2

    00mg-BIDdo

    se(1.8),andhigh

    est

    forthe

    400mg-BIDdo

    se(3.1)

    [184]

    Ibup

    rofen,

    naproxen,diclofenac,

    meloxicam

    ,ind

    omethacin,

    piroxicam,

    andmefenam

    icacid

    Non

    e/UKprim

    arycare

    20years

    729,2

    94NSA

    IDusers;

    443,047controls

    Increase

    inther

    elativer

    atefor

    MIw

    ithcumulativea

    nddaily

    dose

    ofibup

    rofen

    anddiclo

    fenac.Higherrisk

    ofMIw

    ithdiclo

    fenacu

    se(relativer

    atio

    =1.2

    1)than

    ibup

    rofen(relativer

    atio

    =1.0

    4)or

    naproxen

    (relativer

    atio

    =1.0

    3)

    [185]

    Etoricoxib

    anddiclo

    fenac

    TheM

    EDAL

    Stud

    y/Multin

    ational

    19.4to

    20.8mon

    ths

    Etoricoxib

    60mg/d;𝑛=6,769

    90mg/d;𝑛=5,012

    Diclofenac𝑛=11,717

    Thethrom

    botic

    CVris

    kHRof

    etoricoxib

    todiclo

    fenac=

    0.96.P

    rolonged

    useo

    feither

    NSA

    IDresultedin

    anincreased

    riskof

    thrombo

    ticCV

    events

    [186]

    Naproxen,

    ibup

    rofen,

    diclo

    fenac,

    celecoxib,androfecoxib

    Non

    e/USA

    6years

    48,566

    patie

    ntsrecently

    hospita

    lized

    form

    yocardial

    infarctio

    n,revascularization,

    orun

    stableangina

    pectoris

    CVDris

    kincreasedwith

    shortterm

    (<90

    days)u

    seforibu

    profen

    with

    incidence

    rateratio

    sof1.67,diclo

    fenac1.86,

    celecoxib1.3

    7,androfecoxib1.4

    6,bu

    tnot

    forn

    aproxen0.88

    [51]

  • 8 Oxidative Medicine and Cellular Longevity

    Table2:Con

    tinued.

    NSA

    IDClinicaltrialn

    ame/locatio

    nDurationof

    study

    Num

    bero

    fpatientsselected

    Effect

    References

    Cele

    coxib,rofecoxib,valdecoxib,

    ibup

    rofen,

    naproxen,diclofenac,and

    indo

    methacin

    Non

    e/USA

    7years

    610,001p

    atients;with

    out

    CVD𝑛=525,249;w

    ithhisto

    ryof

    CVD𝑛=84752

    Rofecoxib(10.91

    events/

    1000

    person

    -years),valdecoxib

    (12.41

    events/

    1000

    person

    -years),and

    indo

    methacin(13.25

    events/

    1000

    person

    -years)increased

    CVDris

    kin

    patie

    ntsw

    ithno

    histo

    ryof

    CVD.

    Rofecoxibuseincreased

    riskof

    cardiovascular

    eventinpatie

    ntsw

    ithCV

    D(30.28

    events/

    1000

    person

    -years)

    [187]

    Ibup

    rofen,

    diclo

    fenac,rofecoxib,

    celecoxib,andnaproxen

    Non

    e/Danish

    popu

    latio

    n9–

    34days

    153,465healthyindividu

    als

    Dosed

    ependent

    increase

    incardiovascular

    eventsdu

    etouseo

    fdiclo

    fenac(HR=1.6

    3),rofecoxib

    (HR=

    2.13)a

    ndcelecoxib(H

    R=2.01)w

    asob

    served

    [188]

    Cele

    coxib,ibup

    rofen,

    andnaproxen

    TheP

    rospectiv

    eRa

    ndom

    ized

    Evaluatio

    nof

    Cele

    coxibIntegrated

    Safety

    versus

    Ibup

    rofenOr

    Naproxen

    (PRE

    CISION)

    trial/M

    ultin

    ational

    2009-ongoing

    20,000

    patie

    ntsw

    ithsymptom

    aticosteoarthritis

    orrheumatoidarthritisat

    high

    riskforo

    rwith

    establish

    edCV

    D

    Thistrialw

    illdeterm

    inethe

    cardiovascular

    safetyof

    theN

    SAID

    s[189]

    ND

    Non

    e/Denmark

    10years

    107,0

    92patie

    ntsw

    ithfirst

    incidenceo

    fHF

    Theh

    azardratio

    ford

    eath

    duetoMIo

    rHFwas

    1.70,1.7

    5,1.3

    1,2.08,1.22,and1.2

    8forrofecoxib,cele

    coxib,ibup

    rofen,

    diclo

    fenac,naproxen,and

    otherN

    SAID

    s,respectiv

    ely

    [190]

    Ibup

    rofen,

    diclo

    fenac,naproxen,

    rofecoxib,celecoxib,valdecoxib,and

    etoricoxib

    Non

    e/Th

    eNetherla

    nds

    4years

    485,059subjectswith

    first

    hospita

    lisationfora

    cute

    myocardialinfarction,

    CV,

    andgastr

    ointestin

    alevents

    AMIrisk

    with

    celecoxib(O

    R2.53),

    rofecoxib(O

    R1.6

    0),ibu

    profen

    (OR1.5

    6),

    anddiclo

    fenac(OR1.5

    1)was

    significantly

    increased.Sign

    ificant

    increase

    inCV

    risk

    with

    currentu

    seof

    individu

    alCO

    X-2

    inhibitorsandtN

    SAID

    s(ORfro

    m1.17to

    1.64).Significantd

    ecreaseinAMIw

    ithcurrentu

    seof

    naproxen

    (OR0.48)

    [191]

    Ibup

    rofen,

    naproxen,diclofenac,

    etod

    olac,celecoxib,rofecoxib

    Non

    e/NorthernDenmark

    10years

    32,602

    patie

    ntsw

    ithfirst

    atria

    lfibrillationor

    flutte

    rand325,918po

    pulatio

    ncontrols

    Increasedris

    kof

    atria

    lfibrillationor

    flutte

    rwas

    40–70%

    (lowe

    stfor

    non-selectiveN

    SAID

    sand

    high

    estfor

    COX-

    2inhibitors).

    [192]

  • Oxidative Medicine and Cellular Longevity 9

    Table2:Con

    tinued.

    NSA

    IDClinicaltrialn

    ame/locatio

    nDurationof

    study

    Num

    bero

    fpatientsselected

    Effect

    References

    ND

    Non

    e/Denmark

    10years

    83,677

    patie

    ntso

    fwhich

    42.3%received

    NSA

    IDs

    Death/recurrent

    MId

    ueto

    NSA

    IDtre

    atment(even

    shortterm)w

    assig

    nificantly

    high

    er.D

    iclofenacp

    osed

    the

    high

    estrisk

    ofallN

    SAID

    s

    [193]

    ND

    INternationalV

    Erapam

    ilTrando

    laprilSTud

    y(INVES

    T)/M

    ultin

    ational

    7years

    882chronicN

    SAID

    users

    and21,694

    nonchron

    icNSA

    IDusers.Patie

    ntsw

    ithhypertensio

    nandclinically

    stablec

    oron

    aryartery

    disease

    Prim

    aryou

    tcom

    elikea

    ll-causem

    ortality,

    nonfatalMI,or

    nonfatalstroke

    and

    second

    aryindividu

    alou

    tcom

    eslik

    eall-c

    ause

    mortality,cardiovascular

    mortality,totalM

    I,andtotalstro

    keoccurred

    atar

    ateo

    f4.4eventsper100

    patie

    nt-yearsin

    thec

    hron

    icNSA

    IDgrou

    p,versus

    3.7eventsper100

    patie

    nt-yearsin

    then

    onchronicN

    SAID

    grou

    p

    [111]

    Cele

    coxib,rofecoxib,ibup

    rofen,

    diclo

    fenac,naproxen,and

    others

    Non

    e/Denmark

    13years(with

    first

    MI)

    128,418patie

    nts(77%

    participated

    inthes

    tudy)

    Incidences

    ofcoronary

    deathor

    nonfatal

    recurrentM

    Iwith

    NSA

    IDsu

    seremain

    unchangedwith

    thetim

    eelapsed

    even

    after

    5years

    [194]

    ND

    Non

    e/dataob

    tained

    from

    REdu

    ctionof

    Atherothrombo

    sisfor

    Con

    tinuedHealth

    (REA

    CH)registry

    which

    inclu

    desp

    atientsfrom

    Latin

    America,North

    America,Eu

    rope,A

    sia,the

    MiddleE

    ast,andAu

    stralia

    4years

    4,420NSA

    IDusers;39,675

    NSA

    IDno

    nusers

    1.16-fold

    high

    erris

    kof

    CVD,M

    Iin

    NSA

    IDusers

    [41]

    MI:myocardialinfarction,

    HF:heartfailure,C

    VD:cardiovasculard

    iseases,C

    BV:cerebrovascular

    diseases,C

    HF:congestiv

    eheartfailu

    re,H

    R:hazard

    ratio

    ,OR:

    oddratio

    ,ND:not

    defin

    ed,R

    R:rateratio

    ,and

    TIA:

    transie

    ntisc

    hemicattack.V

    arious

    clinicaltria

    lssuggestthe

    increasedincidences

    ofCV

    Din

    NSA

    IDusers.Th

    etablelistson

    lythec

    linicaltrialsrepo

    rted

    between2006

    and2014.

  • 10 Oxidative Medicine and Cellular Longevity

    Thrombosis

    Nonselective (inhibits COX-1 and COX-2)naproxen, ibuprofen, ketoprofen

    COX-2 selectivevaldecoxib, celecoxib, rofecoxib

    Apoptosis

    NADPH oxidasesCell signalling pathways affectedLipoxygenases

    Xanthine oxidasesNitric oxide synthases

    (free radicals)

    Acute myocardial infarction, reperfusion injury, heart failure Oxidative stress

    Cardiovascular diseases

    ROS

    Cytochrome p450

    Arachidonic acid metabolism

    Platelet aggregation on the site of atherosclerotic plaque

    rupture on the coronary artery wall of the heart

    NSAIDs

    ↑ Nonsteroidal activated gene- 1↑ p53 ↑ Bax ↓ Bcl-2↑ Mitochondrial cytochrome c↑ Apoptosis inducing factor↓ Akt↓ NF-𝜅B↓ Proteasome activity

    O2∙− , H2O2,∙OH

    ↓ Prostacyclin↑ Thromboxane

    Thromboxane/prostacyclin imbalance

    Figure 2: Pathways involved in the development of cardiovascular diseases by NSAIDS. The figure shows the upregulation anddownregulation of various pathways by NSAIDs leading to the development of CVD. Mitochondria play a major role in the generationof ROS induced by NSAIDs followed by oxidative stress and finally CVD.

    found to increase the risk of atherothrombosis [46]. TheCOX-2 selective NSAID rofecoxib at low doses (50mg daily)increased the occurrences of myocardial infarction by 0.5%in approximately 8000 patients with rheumatoid arthritiscompared to 0.1% in those treated with naproxen (500mgtwice daily), indicating a severe thrombogenic effect of rofe-coxib compared to naproxen [47].Themain increased risk ofcardiovascular events associated with COX-2 inhibitors wasan increased risk of myocardial infarction [46].

    The report of Adenomatous Polyp Prevention on Vioxx(APPROVe) trial on the adverse effect of rofecoxib on CVDultimately led to the discontinuation of this drug in severalcountries [48]. Increased thromboembolic events have beenassociated with rofecoxib compared to naproxen [49]. Inanother trial rofecoxib was found to be associated withcardiac arrhythmias and renal conditions [50]. Howeversimilar adverse effects were not encountered in patientstreated with other COX-2 inhibitors [50]. More importantly,several semiselective NSAIDs like diclofenac and meloxicamand nonselective NSAIDs including naproxen and ibuprofenhave also been shown to increase the incidences of CVD[46, 51].Of these drugs, diclofenac has been shown to increase

    the occurrences of myocardial infarction and stroke evenat lower doses of 1000mg/day [51]. This effect of diclofenac hasbeen attributed to its greater selectivity for COX-2 inhibition.It has also been reported that ibuprofen is associated withCVD comparable to the effect of COX-2 selective inhibitor,celecoxib [20]. It has been suggested that the differencebetween the effect exhibited by rofecoxib and other NSAIDsof the same class on cardiovascular incidences is due tothe distinct chemical properties and prooxidant activity ofrofecoxib [52]. The toxic effect of rofecoxib was reportedlydue to its ability to reduce the low density lipoprotein (LDL)antioxidant capacity as a result of increased lipid peroxidation[52]. Merck voluntarily withdrew rofecoxib (Vioxx) in 2004.Pfizer was asked by the US Food and Drug Administration(FDA) to withdraw valdecoxib (Bextra) from the market in2005 because of a higher than expected number of reportsof serious and potentially life-threatening skin reactions anddeaths.

    Naproxen seems to show less cardiovascular events thanother commonly used NSAIDs, possibly because it mimicsthe activity of acetylsalicylic acid (aspirin) by suppressing

  • Oxidative Medicine and Cellular Longevity 11

    cyclooxygenase platelet thromboxane B2 [53]. However, therole of naproxen in the development of CVD has been con-troversial. Several studies have reported the cardioprotectiveeffect of the compound [54–56], and on the other handincreased cardiovascular risk has been associatedwith the useof this NSAID [57–59]. The only NSAID which has not beenassociated with increased cardiovascular events is aspirin.Aspirin is the only known NSAID which has antithromboticactivity through the inhibition of platelet aggregation inthe artery of the heart thereby exhibiting cardioprotectiveeffects [60]. It does so by acetylating the platelet COX-1and irreversibly suppressing thromboxane A2 production,which is required for platelet aggregation and thrombusformation [60]. Naproxen on the other hand causes reversibleinhibition of cyclooxygenase by binding to the enzyme [61].It was found that inhibition in thromboxane A2 synthesis wasmore pronounced (75%) compared to prostacyclin inhibition(50%) after the oral administration of 500mg naproxen inhealthy volunteers [61].

    5. NSAIDs and Reactive OxygenSpecies Generation

    NSAIDs have been shown to be associated with increasedROS production (Table 3). In the heart the main producer ofROS is the mitochondria [62]. Under normal physiologicalconditions, mitochondria generate ROS as a consequence ofaerobic respiration [63]. During aerobic respiration about 5%of O2consumed via aerobic reaction is converted into ROS

    [63]. The mitochondria-dependent overproduction of ROShas been reported under numerous pathological conditionsincluding myocardial heart failure, inflammatory diseases,cancer, hypertension, and diabetes [64–67]. In myocardialheart failure, cardiomyocytes have been shown to be targetedby excessive ROS generation [64].

    ROS levels and the redox state of a cell are consideredto be important in the dysfunction of various biologicalsignaling pathways. The formation of ROS via the reductionof molecular oxygen or by the oxidation of water leads to theformation of free radicals such as superoxide anion (O

    2

    ∙−),hydroxyl radical (∙OH), and hydrogen peroxide (H

    2O2).

    Oxidative stress arises when the oxidant production (sum ofall the ROS) surpasses the antioxidant capacity in the cells.Under normal physiological conditions low amounts of O

    2

    ∙−

    in the cardiomyocytes are converted to the less toxic H2O2

    by the enzyme superoxide dismutase (SOD). The reactionfurther proceeds by the formation of water by the action ofthe enzyme catalase or glutathione peroxidase (GPx) system[68]. However, when a homeostatic imbalance between thecellular antioxidant capacity and ROS levels occurs, elevatedROS levels can damage cellular macromolecules includinglipids, proteins, and nucleic acids. High levels of ROS alsoaccelerate cell death due to apoptosis as well as necrosis by theactivation of poly(adenosine diphosphate ribose) polymerase[69] and thus significantly contribute to the development ofvarious pathological conditions [70].

    The generation of ATP in the mitochondria utilizes theelectrons from reduced substrates that are transferred to anacceptor molecule of the electron transport chain (ETC).

    Leakage of electrons from the mitochondria ETC results inO2

    ∙− formation [71]. Electron leakage is potentially possibleat 9 sites in the ETC; however most of the ROS seem tobe associated with complexes I and III which have beenwell studied [72]. The generation of ROS increases in intactmitochondria as well as in submitochondrial particles dueto the oxidation of complex I substrates as a result ofinhibiting complex III by antimycin A [72]. On the otherhand, rotenone, an inhibitor of complex I, prevented theantimycin A induced ROS generation in mitochondria butnot in the submitochondrial particles [72].

    The ROS status in the cellular system regulates manybiological processes.While increased levels of ROS have beenshown to be involved in various pathological conditions,under basal conditions, the generation of free radicals in theheart is needed for cellular responses including regulatingmyocyte growth and maintaining vascular smooth muscletone [73]. Basal levels of ROS play an important role in theincrease of cell cycle progression and intracellular signalingassociated with phosphorylation of several signaling proteinslike mitogen-activated protein kinases (MAPKs) and proteinkinase B [74]. Under normal physiological conditions, ROSupregulates the Akt signaling pathway and promotes cellsurvival [75]. ROS also behaves as second messengers in sig-naling pathwayswherein it has been demonstrated that, in thepresence of basal ROS levels, tyrosine phosphatase activityis higher relative to its kinases [76]. Ligand stimulation (asin the case of neutrophils, through the binding of cytosolicproteins to a membrane bound oxidase) leads to increasedROS levels and deactivation of tyrosine phosphatases with aconsequent increase in the kinase activity [76].This conditionis transient and is reversed by reductions in ROS levels. ROShas also been demonstrated to be involved in the modulationof transcription factors like NF-𝜅B, Hif-1 [77], and severalcardiac transcription factors like SRF, Sp1, AP-1, GATA-4, andMEF2C [78].

    Current experimental data suggest that the main mecha-nism throughwhichNSAIDs exert their anticancer activity isthrough the generation of ROS leading to oxidative stress andfinally apoptosis in cancer cells [79–82]. ROS is accompaniedby the activation and inhibition of several signaling pathwaysassociated with cell death and cell survival although contro-versies exist regarding the role of ROS in the downregulationand upregulation of these pathways [83, 84].TheAkt pathwayis one of the most important pathways for promoting cellsurvival and growth, and it has been shown that high ROSlevels are lethal and can inactivate the Akt signaling pathway[85]. NF-𝜅B is an important transcription factor involved incell survival, inflammation, and stress responses. Its down-regulation was demonstrated by exposure of HLEC (humanlymphatic endothelial cells) cells to sustained oxidative stress[86]. Increased levels of H

    2O2caused by addition of glucose

    oxidase to HLECs prevented NF-𝜅B(p65) regulated geneexpression by blocking translocation of NF-𝜅B to the nucleus[86]. NF-𝜅B regulated gene expression is important for abroad range of physiological processes [86].

    Sulindac, a nonselectiveNSAID, as well as itsmetabolites,generates ROS in different cancer cell lines [80–82]. Similarly,diclofenac-induced apoptosis of various kinds of cancer cells

  • 12 Oxidative Medicine and Cellular Longevity

    Table3:NSA

    IDsind

    uced

    ROSgeneratio

    nin

    different

    celltypes.

    (a)

    NSA

    IDs

    Non

    selective/semise

    lective

    Sourceso

    fROSgeneratio

    nCells/mod

    els/anim

    alsstudied

    Outcomes

    References

    Diclofenac,indo

    methacin,

    ketoprofen

    Mito

    chon

    drialrespiratio

    nSaccharomyces

    cerevisia

    eYeast

    cells

    BY4741

    andmito

    chon

    drial

    DNAdeletio

    n(rho

    0 )str

    ains

    TheseN

    SAID

    stargetm

    itochon

    driato

    indu

    cecelltoxicity

    [95]

    Ibup

    rofenandnaproxen

    TheseN

    SAID

    Sindu

    cetoxicityindepend

    ento

    fmito

    chon

    drialrespiratio

    n[95]

    Indo

    methacin,

    sodium

    diclo

    fenac,flu

    rbiprofen,

    zalto

    profen,and

    mofezolac

    ND

    Hum

    angastric

    epith

    elialcellline

    AGS

    AllNSA

    IDse

    xceptm

    ofezolac

    increased

    apop

    totic

    DNAfragmentatio

    nandexpressio

    nof

    COX-

    2mRN

    A.D

    NAfragmentatio

    nindu

    ced

    byindo

    methacinor

    flurbiprofenwas

    redu

    ced

    byantio

    xidants.Indo

    methacinat1m

    Mwas

    apo

    tent

    indu

    cero

    fROSgeneratio

    nin

    thec

    ells

    [79]

    Diclofenaca

    ndnaproxen

    NADPH

    oxidases

    Spon

    taneou

    shypertensiver

    ats

    NADPH

    oxidasee

    xpressionincreasedin

    the

    heartand

    aortab

    ydiclo

    fenaca

    ndnaproxen.

    ROSlevelsincreaseddu

    etoNADPH

    oxidase

    [114]

    Hum

    anEA

    .hy926

    Endo

    thelialcells

    Nox2iso

    form

    ofNADPH

    oxidaseisincreased

    bydiclo

    fenac

    [114]

    Aspirin,

    naproxen,and

    piroxicam

    NADPH

    oxidases

    Ratadipo

    cytes

    Activ

    ationof

    NOX4iso

    form

    ofNADPH

    oxidaser

    esultsin

    theg

    enerationof

    H2O

    2[115]

    Indo

    methacin

    Xanthine

    oxidases

    Hum

    ancolonica

    deno

    carcinom

    acells

    (Caco-2cells)

    Xanthine

    oxidasea

    ctivity

    increasesb

    ymore

    than

    100%

    1hou

    rafte

    rtreatment

    [96]

    Indo

    methacin

    Mito

    chon

    driasuperoxide

    leakage

    Ratgastricepith

    elialcellline

    RGM1

    Indo

    methacinindu

    cedtheleakage

    ofsuperoxide

    anionin

    theisolatedmito

    chon

    dria

    from

    theg

    astricRG

    M1cells

    [93]

    Indo

    methacin,

    diclo

    fenac

    sodium

    ,and

    aspirin

    ND

    Ratgastricepith

    elialcellline

    RGM1and

    ratsmallintestin

    alepith

    elialcelllineIEC

    6

    Indo

    methacin,

    diclo

    fenac,andaspirin

    ingastric

    RGM1cellsandsm

    allintestin

    alIEC6

    cells

    increasedlip

    idperoxidatio

    n[93]

    Sulin

    dac,sulin

    dacs

    ulfid

    e,andsulin

    dacs

    ulfone

    ND

    Pancreas

    carcinom

    aBXP

    C3,

    glioblastomaA

    172,colon

    carcinom

    aDLD

    -1,oral

    squamou

    sSAS,andacute

    myelocytic

    leuk

    emiaHL6

    0cells

    TheN

    SAID

    sulin

    daca

    ndits

    metabolites

    sulin

    dacs

    ulfid

    eand

    sulin

    dacs

    ulfone

    all

    increasedRO

    Sgeneratio

    n.Sulin

    dacs

    ulfid

    eshow

    edtheg

    reatestR

    OSgeneratio

    n

    [80]

    Indo

    methacin,

    piroxicam,

    andaspirin

    Lipo

    xygenases

    Gastricandintestinalmucosain

    mice

    Results

    inan

    overprod

    uctio

    nof

    leuk

    otrie

    nes

    andprod

    uctsof

    5-lip

    oxygenasea

    ctivity.

    Increasesinleuk

    otrie

    neand5-lip

    oxygenase

    activ

    ityhave

    been

    associated

    with

    ROS

    generatio

    n

    [123,124,127]

    Indo

    methacin

    Lipo

    xygenases

    Efferentgastriccirculationofpigs

    Timed

    ependent

    form

    ationof

    leuk

    otrie

    ne-C

    4[128]

  • Oxidative Medicine and Cellular Longevity 13

    (a)Con

    tinued.

    NSA

    IDs

    Non

    selective/semise

    lective

    Sourceso

    fROSgeneratio

    nCells/mod

    els/anim

    alsstudied

    Outcomes

    References

    Diclofenac

    CytochromeP

    450enzymes

    Saccharomyces

    cerevisia

    eyeast

    cells

    expressin

    gthem

    utantP

    450

    BM3M11(capableof

    metabolizingdiclo

    fenacs

    imilar

    tohu

    mans)

    IncreasedRO

    Sprod

    uctio

    nby∼1.5

    and1.8

    times

    atconcentrations

    of30𝜇M

    and50𝜇M,

    respectiv

    ely,com

    paredto

    wild

    type

    [140]

    Diclofenac,indo

    methacin,

    ketoprofen,and

    naproxen

    CytochromeP

    450enzymes

    Saccharomyces

    cerevisia

    eYeast

    cells

    BY4741

    andmito

    chon

    drial

    DNAdeletio

    n(rho

    0 )str

    ains

    NSA

    IDsm

    etabolism

    associated

    with

    increased

    P450-related

    toxicity

    [95]

    (b)

    NSA

    IDs

    coxibs

    Sourceso

    fROSgeneratio

    nCells/mod

    els/anim

    alsstudied

    Outcomes

    References

    Ibup

    rofen,

    ketoprofen,and

    indo

    methacin

    CytochromeP

    450enzymes

    Bacillusm

    egaterium

    AlltheN

    SAID

    scausedam

    arkedincrease

    inthetotalcytochromeP

    450level

    [142]

    Aspirin

    ,diclofenac,diflu

    nisal,

    flurbiprofen,

    ibup

    rofen,

    indo

    methacin,

    ketoprofen,m

    efenam

    icacid,n

    aproxen,

    piroxicam,sulindac,andtolm

    etin

    CytochromeP

    450enzymes

    Rath

    epatocytes

    Cytotoxicityof

    diclo

    fenac,ketoprofen,

    andpiroxicam

    was

    increasedby

    cytochromeP

    450causinghepatotoxicity

    [138]

    Diclofenaca

    ndnaproxen

    Endo

    thelialn

    itricoxide

    synthase

    Spon

    taneou

    shypertensiver

    ats

    Increasedexpressio

    nof

    eNOSmRN

    Adu

    eto

    theg

    enerationof

    H2O

    2which

    isrespon

    sibleforu

    pregulationof

    eNOSat

    thetranscriptio

    naland

    post-

    transcrip

    tionallevels

    [114]

    Rofecoxibandcelecoxib

    NADPH

    oxidases

    Spon

    taneou

    shypertensiver

    ats

    NADPH

    expressio

    nincreasedin

    theh

    eart

    andaortab

    yrofecoxibandcelecoxib

    [114]

    Hum

    anEA

    .hy926

    Endo

    thelialcells

    Nox2Ex

    pressio

    nincreasedby

    rofecoxib

    [114]

    Rofecoxibandcelecoxib

    Endo

    thelialn

    itricoxide

    synthase

    Spon

    taneou

    shypertensiver

    ats

    Inthea

    orta,the

    coxibs

    didno

    tsho

    wany

    eNOSmRN

    Aexpressio

    n.In

    theh

    eart

    onlyrofecoxibshow

    edas

    ignificant

    increase

    inthee

    xpressionof

    eNOS

    [114]

    Etod

    olac

    ND

    Hum

    angastr

    icepith

    elialcell

    lineA

    GS

    Increasedapop

    totic

    DNAfragmentatio

    nandexpressio

    nof

    COX-

    2mRN

    A[79]

    Nim

    esulide

    NADPH

    oxidases

    Ratadipo

    cytes

    Activ

    ationof

    NOX4iso

    form

    ofNADPH

    oxidaser

    esultsin

    theg

    enerationof

    H2O

    2[115]

    * Alth

    ough

    otherreportsarea

    vailables

    uggesting

    theroleo

    fNSA

    IDsinRO

    Sform

    ation,thetablelistson

    lythoseN

    SAID

    sfor

    which

    results

    show

    thattheseN

    SAID

    sareassociated

    with

    CVDor

    NSA

    IDsm

    entio

    ned

    inthetext.

  • 14 Oxidative Medicine and Cellular Longevity

    has been reported [87]. This apoptosis is mainly mediatedby an increase in the intracellular levels of the ROS [87]. Inthe cardiovascular system the major sources of ROS gener-ation include the mitochondria, NADPH oxidases, xanthineoxidoreductases, lipoxygenase, cyclooxygenases, nitric oxidesynthases, and cytochrome P450 based enzymes (Figure 2).Continuous exposure of cardiovascular cells to oxidativestress associated with elevated ROS levels would result inaltered cellular homeostasis which could be an importantcontributing factor for various cardiovascular conditions.Endothelial cells play a critical role in maintaining vas-cular homeostasis which is important for the control ofmany cardiovascular diseases including atherosclerosis andthrombosis [88]. In human umbilical vein endothelial cells(HUVEC), 160 𝜇M sulindac induced endothelial apoptosisas determined by a time dependent increase in annexin Vpositive cells. Both sulindac and indomethacin significantlyincreased cleaved poly(ADP-ribose) polymerase (PARP) lev-els as well as increasing the level of the apoptotic activatingfactor caspase-3 [89].The apoptosis in HUVEC cells inducedby the NSAIDs was associated with reduced PPAR𝛿 and 14-3-3-𝜀 expression. Under normal conditions 14-3-3-𝜀 bindsto phosphorylated Bad inhibiting translocation of Bad tothe mitochondria and preventing apoptosis through themitochondrial pathway. However, sulindac through the sup-pression of 14-3-3-𝜀 expression increased Bad translocationto mitochondria thereby inducing apoptosis [89].

    6. Mitochondria Are the Main TargetOrganelles of the NSAIDs

    It has been shown that the heart is more susceptible toROS generation induced by drugs like doxorubicin comparedto other tissues of the body although the drugs are evenlydistributed throughout the body [90]. This is possibly dueto high levels of mitochondria in the heart which are themajor producers of ROS in the cardiovascular system. Over90% of ATP required for the normal functioning of theheart is provided by the mitochondria, which utilizes anefficient oxidative phosphorylation system. ATP productionmay increase depending upon the requirements of the body,especially at times of excessive physical exertion or other hor-monal stimulations [91].Mitochondria are not only themajorproducers of the free radicals [72], but excessive generationof ROS in turn targets the mitochondria itself [92]. NSAIDshave been shown to have adverse effects on the mitochondriaresulting in the increased production of ROS [93, 94]. In yeastcells different NSAIDs generated ROS which was associatedwith delayed growth in wild-type cells [95]. Yeast cells lackingmitochondrial DNA were resistant to a delay in cell growth[95]. More specifically the yeast deletion strains lacking thegenes encoding subunits of the mitochondrial complexes IIIand IV were significantly resistant to diclofenac as well asindomethacin and ketoprofen [95]. This data suggests thatmitochondria are the main target organelles through whichthese compounds exert their toxic effect on these cells. Thedata also suggest that mitochondrial complex III and/or

    complex IV are affected by NSAIDs resulting in increasedROS production.

    The nonselective NSAID indomethacin has been shownto target mitochondria by directly inhibiting mitochondrialcomplex 1 of human colonic adenocarcinoma cells. Theinhibition of complex I was accompanied by a decrease in theintracellular ATP levels within 10–30 minutes [96]. Studiesdirectly related to mitochondrial dysfunction or damage inthe presence of NSAIDs in CVD have not been carriedout but are needed (Table 2). Although the mechanismby which NSAIDs can cause mitochondrial dysfunction incardiomyocytes is not fully understood, preventing increasedmitochondrial ROS levels will reduce the adverse effects ofelevated ROS levels and may reduce the incidences of CVDcaused by NSAIDs.

    Apart from the role of NSAIDs in causing cardiovascularincidences, NSAIDs affect other cardiovascular (CV) param-eters including left ventricular function, infarct size, andblood pressure.In elderly patients (71.8 ± 7.6 years) NSAIDexposure for 14 days werefound to have higher left end-diastolic dimension (+1.96mm)but no significant changes in other echocardiographic param-eters compared to non-NSAID users [97].

    The impact of NSAIDs on infarct size during myocardialinfarction has only been investigated by a few laboratories.Indomethacin (10mg/kg) was found to increase myocardialinfarct size in animals, while ibuprofen (6.25mg/kg/h) hadthe opposite effect [98–100]. The authors suggested that theopposite effect on the infarct size could be attributed tovariable doses of the NSAIDs, different degrees of inhibitionof prostaglandin and its by-products, and others factorslike myocardial oxygen consumption [99]. The same groupreported that the frequency of acute infarct expansion syn-drome in patients with symptomatic pericarditis treated withindomethacin was 22% compared to ibuprofen which wasonly 8% [101].Thedegree of infarct expansionwas also greaterin patients treated with indomethacin compared to ibuprofen[101].

    Another important CV parameter is hypertension, whichis one of the major contributors to the development of CVD.A series of events occur in patients with prolonged highblood pressure including left ventricular hypertrophy, systolicand diastolic dysfunction leading to arrhythmias, and heartfailure [102]. Except for aspirin, all NSAIDs could potentiallyincrease blood pressure when taken at doses necessary toalleviate pain and inflammation in both hypertensive andnormotensive individuals [39].

    NSAIDs through their ability to block COX enzymeslead to the inhibition of renal prostaglandin [103]. Renalprostaglandins modulate several renal functions whichinclude maintaining renal homeostasis and exerting diureticand natriuretic effects [104, 105]. Inhibition of the natriureticeffect of COX-2 leads to an increase in sodium retentionthereby leading to excess water retention in humans [106].The inhibition of renal vasodilating prostaglandins induces

  • Oxidative Medicine and Cellular Longevity 15

    the production of vasoconstricting factors like vasopressinand endothelin-1.This also results in water retention, therebyleading to an increase in the total blood volume and caus-ing altered systolic and diastolic blood pressure [103, 106,107]. NSAIDs like ibuprofen, indomethacin, and naproxenincrease the mean arterial pressure by 5 to 6mmHg inpatients with established hypertension [108, 109], which maybe enough to raisemedical concerns under certain conditions[110]. Additionally, the efficiency of all antihypertensivemedications except for calcium channel blockers may be sub-stantially reduced byNSAIDs [40]. SinceCVoutcomeswouldhave different pathogenesis, increased ROS levels may notbe the underlying mechanism for some occurrences of CVD.Other CV related parameters (such as hypertension), whichare important for the normal functioning of the heart, are alsolikely to affect the CV outcome. Although direct comparisonsbetween the different clinical studies shown in Table 2 are notpossible, these studies suggest that CVD in NSAID users areinfluenced by the study populations used (such as with versuswithout preexisting acute myocardial infarction). Chronicuse ofNSAIDs by hypertensive individuals has been shown toincrease the incidences of myocardial infarction, stroke, andcardiovascular mortality compared to nonchronic NSAIDusers [111] (Table 2).

    7. NSAIDs and Nicotinamide AdenineDinucleotide Phosphate-Oxidase

    Apart frommitochondria being themajor source of ROS gen-eration, the plasma membrane bound nicotinamide adeninedinucleotide phosphate-oxidase (NADPH oxidase) has alsobeen indicated in the production of ROS in phagocytes likeneutrophils andmacrophages where it produces an “oxidativeburst” ofO

    2

    ∙− [112].NADPHoxidases are considered to be themajor source of nonmitochondrial ROS generation in manycells. The formation of free radicals is due to the electrontransferred from theNADPH tomolecular oxygen during theprocess of phagocytosis. The phagocytic NADPH oxidasesplay an important role in the host defensemechanism and areinvolved in killing pathogens ingested during phagocytosis.Apart from the phagocytic NADPH oxidase, in the lastdecade NADPH oxidase-dependent ROS generation was alsoidentified in nonphagocytic cells including the endothelialcells, vascular smooth muscle cells, and cardiomyocytes ofthe cardiovascular system [113]. Compared to the phago-cytic NADPH oxidases, the nonphagocytic enzymes producelower amounts of ROS continuously and these levels mayincrease in the presence of specific extrinsic stimuli [113].More interestingly, the ROS generated by NADPH oxidaseslead to an enhancement in levels of ROS produced fromother sources [112]. In one such study, mitochondria andNADPHoxidase 1 isoenzyme (Nox 1) were found to be closelycoordinated for sustained generation of ROS leading tooxidative stress and cell death [112].When human embryonickidney 293T cells were exposed to serum-free media elevatedROS levels occur within 5 minutes of exposure and persistedfor 8 hrs [112]. Utilizing RNA interference Nox isoenzymeswere demonstrated to play a role in the induction of serum-withdrawal ROS generation. Although low levels of Nox 2

    and Nox 4 did not affect the generation of serum-withdrawalinduced ROS generation, low levels of Nox 1 led to a decreasein the ROS formation in these cells at 8 h (late phase). Usingdifferent mitochondrial complex inhibitors such as rotenoneand potassium cyanide (KCN) mitochondria were found toplay a role in the early phase (first 30 minutes) of ROSgeneration in the cells exposed to serum-freemedia [112].Theauthors of the study suggested that mitochondrial generationof ROS occurs first, followed by the action of Nox 1 in theserum deprived cell system [112].

    The adverse effect of NSAIDs on the cardiovascularsystemwas evident by a study which reported that whenmalespontaneously hypertensive rats (SHR) were treated eitherwith coxibs or with nonselective NSAIDs like diclofenacand naproxen, the mRNA expression of NOX enzymes 1, 2,and 4 was markedly increased in the heart and aorta [114].Furthermore the oxidative stress in the heart and aorta ofthe NSAID treated animals was also elevated along with anincrease in the O

    2

    ∙− production. Of all the NSAIDs used,diclofenac was the most potent inducer of NADPH oxidases[114]. The role of NADPH oxidases in the generation ofROS in the animals was ascertained by studying the effectof apocynin (an NADPH oxidase inhibitor) in the reversalof the oxidative stress induced by diclofenac [114]. Activationof Nox 4 by NSAIDs like aspirin, naproxen, nimesulide, andpiroxicam has also been reported in rat adipocytes resultingin higher production of H

    2O2[115].

    8. NSAIDs and Xanthine Oxidase

    Xanthine oxidoreductase (XOR) is a conserved molybd-oflavoenzyme occurring in milk and some tissues includ-ing the liver, heart, and small intestine [116]. It has twointerconvertible forms: xanthine dehydrogenase (XDH) andxanthine oxidase (XO). Both enzymes catalyze the conversionof hypoxanthine to xanthine and xanthine to uric acid [116].Although both enzymes are involved in purine degradation,XO is involved only in the reduction of oxygen while XDH isinvolved in the reduction of both oxygen and to a larger extentNAD+.The enzymeXDHhas the ability to bind toNAD+ and,following the oxidation of hypoxanthine to xanthine, reducesNAD+ to NADH. On the other hand, XO, not being able tobind to NAD+, produces the free radical, O

    2

    ∙− by the transferof electrons to molecular oxygen. Apart from O

    2

    ∙−, H2O2

    was also shown to be a major free radical generated by XO[117]. XDH can utilize molecular oxygen and generate ROSbut to a lesser extent than XO. Compared to the other sourcesof ROS generation like mitochondria and NADPH oxidases,the role of XOR as a producer of ROS has been previouslyoverlooked due to its relatively low activity in the heart ofanimals and human. Recently the role of XOR in mediatingvarious pathological conditions has been demonstrated in thecardiovascular system due to significant increases in cellularXOR levels [118]. This was evident by the inhibition of XORby compounds like allopurinol and oxypurinol which lead toa decrease in the oxidative tissue damage. Apart from theirproperty of inhibiting XOR, allopurinol and oxypurinol havealso been shown to act as free radical scavengers and inhibitlipid peroxidation as well as inducing heat shock proteins

  • 16 Oxidative Medicine and Cellular Longevity

    during times of oxidative stress [118]. Interestingly, a highlevel of serum uric acid, the end product of XOR, has beenshown to be a marker for impaired oxidative metabolism[119].

    Although the prevalence of this enzyme has beenreported in human hearts [116, 120] and although increasedlevels of uric acid in myocardial heart failure have beenreported in the literature [119], the role of NSAIDs on XOin the development of various cardiovascular conditions hasnot been investigated. In a study to investigate the adverseeffect of aspirin on gastric mucosal lining leading to pepticulcers and intestinal bleeding, rat gastric mucosal cells wereincubated with aspirin and cytotoxicity already induced byhypoxanthine/XO was determined [121]. Aspirin was shownto increase the cytotoxicity induced by XO. This increase incytotoxicity in the presence of hypoxanthine/XO was incor-porated with an increase in uric acid levels which suggestsROS generation in these cells. Aspirin had an additive effect tothese changes induced by hypoxanthine/XO [121]. In anotherstudy, it was seen that indomethacin augmented the XOactivity in human colonic adenocarcinoma cells bymore than100% upon 60 minutes of exposure which resulted in a timedependent increase in the rate of lipid peroxidation [96].

    9. NSAIDs and Lipoxygenase

    Arachidonic acid that is formed by the action of diacylglyc-erol and cytosolic phospholipase A2 (cPLA

    2) from the mem-

    brane phospholipids is a substrate involved in the formationof either prostaglandins and thromboxanes or leukotrienes bythe cyclooxygenase and lipoxygenase pathway respectively.The oxidation of arachidonic acid by these enzymes leadsto the formation of ROS as the by-product [122]. It wasshown that cPLA

    2-arachidonic acid linked cascade could

    lead to an increase in ROS generation via the lipoxygenasepathway in the rat fibroblasts [123]. The same group reportedthat the generation of ROS triggered by tumor necrosisfactor- (TNF-) 𝛼 was mediated by the activation of cPLA

    2

    and the metabolism of arachidonic acid by 5-lipoxygenase[124]. Lipoxygenase has been reported to be involved inthe upregulation of NADPH oxidases and increased ROSgeneration [125].

    While NSAIDs are well known to inhibit the cyclooxyge-nase pathway, they do not inhibit the formation of leukotri-enes by the lipoxygenase pathway. NSAIDs increases arachi-donic acid levels [82, 126] and arachidonic acid itself canincrease ROS generation [82]. Increased levels of leukotrienesand other metabolites of the lipoxygenase pathway inducedby NSAIDs lead to gastric mucosal damage [127]. Theeffect of NSAIDs like indomethacin, piroxicam, and aspirinon the development of gastric lesions could be reversedby 5-lipoxygenase inhibitors and leukotriene antagonists[127]. This is consistent with NSAIDs increasing the lev-els of leukotrienes and products of 5-lipoxygenase activ-ity. Increased occurrences of leukotriene C4 productionwere also observed in the gastric circulation induced byindomethacin [128]. The gastric and intestinal lesions inthe presence of indomethacin could be reverted by 5-lipoxygenase inhibitor. Increased gastricmucosal leukotriene

    B4 production was observed in patients suffering fromarthritis due to prolonged NSAID treatment [129]. NSAIDshave also been reported to increase the expression of 15-lipoxygenase-1 protein in colorectal cancer cells resulting inthe inhibition of cell growth and apoptosis [130]. Althougha growing body of evidence supports the idea that NSAIDsupregulate the lipoxygenase pathway, no direct evidence isavailable on the precise biochemical mechanism by whichNSAIDs induced lipoxygenase pathway activity increase therate of ROS generation. It is possible that the upregulation ofthe lipoxygenase pathway by NSAIDs may contribute to thenet increase in free radicals leading to apoptosis and oxidativecell damage and ultimately cell death.

    10. NSAIDs and Cytochrome P450

    Cytochrome P450 (P450) is a family of biotransformationenzymes involved in critical metabolic processes [131]. Theyare a class of more than 50 enzymes that catalyze a diverseset of reactions with a wide range of chemically dissimilarsubstrates [131]. In humans they are primarily membrane-associated proteins that can be found in the inner mem-brane of the mitochondria or endoplasmic reticulum of cell[132]. They are predominantly expressed in the liver but arepresent in other tissues of the body as well.This group ofproteins belongs to the heme-thiolate enzyme family, andthey are involved in the oxidative metabolism of numerouscompounds of endogenous and exogenous origin [133]. Therole of P450 in drug metabolism has been considered tobe a key factor in overcoming the adverse and toxicologicaleffects of drugs [134]. The P450 system has been shown toplay an important role in activation of oxygen and ROSgeneration [135, 136]. Normally, the active oxygen species areformed in situ during the P450 cycle when it reacts with asubstrate. However, uncoupling of the P450 system results inexcessive ROS generation and the P450 system being unableto metabolize a substrate, which leads to oxidative stress andsubsequently cellular damage [133, 137].

    Several studies have indicated the role of P450 intoxicity induced by NSAIDs [138–140]. One such NSAID,diclofenac, has been associated with hepatotoxicity due topoor metabolism by P450 [140]. Expression of a drug metab-olizing mutant of P450, BM3M11, in yeast mimicked theoxidative metabolite profiles of diclofenac metabolized byhumanP450s [141].The treatment of the yeast cells expressingthe mutant BM3M11 with 30 𝜇M and 50𝜇M diclofenacincreased the ROS production in the cells significantly by 1.5and 4 times, respectively, compared to cells not treated withthe compound [140]. In another study by the same group,it was shown that diclofenac as well as indomethacin andketoprofen showed an increase in ROS generation by 1.5–2times compared to control [95]. On the other hand, P450 hasbeen shown to be upregulated in bacterial cells by NSAIDslike ibuprofen, ketoprofen, and indomethacin by 11.8-fold,3.9-fold, and 3.0-fold, respectively, relative to control cells[142]. Although no direct evidence is yet available on theinvolvement of P450 in the generation of ROS by NSAIDsin CVD, experimental data available in other cells suggest

  • Oxidative Medicine and Cellular Longevity 17

    that NSAIDs may lead to increased ROS levels via theupregulation of P450 in cardiovascular related cells.

    11. NSAIDs and Nitric Oxide Synthases

    Nitric oxide (NO) is among the few gaseous biological mes-sengers and can be synthesized from l-arginine by endothelialnitric oxide synthase (eNOS). NO has emerged as a keysignaling molecule for maintaining vasodilation and thedysfunction of enzymes associated with NO production hasbeen implicated in various pathological conditions includingCVD, diabetes, and hypertension. When eNOS, which isresponsible for regulating vascular homeostasis, is impaireddue to a lack of the cofactor tetrahydrobiopterin (BH4), itresults in the generation of O

    2

    ∙− rather than NO [143]. Thisstate is referred to as the uncoupled state of eNOS and ismainly attributed to the deficiency or lack of BH4 leading tothe generation of free radicals [144].The depletion of the BH4can be the result of severe oxidative stress.

    A 1.5-fold decrease in plasma nitrite levels was demon-strated in SHR treated with nonselective COX inhibitorsand coxibs [114]. Although earlier reports demonstrated acardioprotective effect of naproxen, a twofold decrease inplasma nitrite levels was observed after treatment of SHRwith naproxen suggesting diminished levels of bioactive NO[114]. Additionally, the authors reported an upregulation inthe eNOS mRNA expression levels rather than a decrease bydiclofenac and naproxen by >2-fold. This increase has beenattributed to the generation of H

    2O2which increases eNOS

    expression at the transcriptional and posttranscriptionallevels [145]. Diclofenac treated animals showed an increasein O2

    ∙− production which could be inhibited by the NOSinhibitor l-NAME. NSAIDs have also been shown to impairthe NO induced vasodilation in healthy individuals possiblydue to increased oxidative stress induced by NSAIDs due tothe uncoupling of eNOS [114].

    12. NSAIDs, ROS, and Cardiovascular Diseases

    Li et al. investigated the effect of several NSAIDs onfree radical generation, NADPH oxidase expression, eNOSexpression, and nitrite levels in the rat aorta and heart usinga mouse model of hypertension as well as vasodilation inhuman subjects [114]. As previously described in the sectionsNSAIDs and nicotinamide adenine dinucleotide phosphate-oxidase and NSAIDs and nitric oxide synthases, this studydemonstrated that the heart and aorta of the NSAID treatedanimals showed increased O

    2

    ∙− production and oxidativestress.Theoxidative stresswas due toROS formation bymanyenzymes including NADPH oxidases, with diclofenac beingthe most potent inducer of NADPH oxidases [114].

    Rat embryonic H9c2 cardiac cells exposed to celecoxib,at concentrations of 10 𝜇M and 100 𝜇M, demonstrated adecrease in cell viability by 45% and 92%, respectively, andthis was associated with a decrease in the expression levelsof the cell survival protein Bcl2 [146]. However, 1 𝜇M ofcelecoxib had no significant effect on cell viability and wasassociated with an upregulation of the Bcl

    2transcript level

    by ∼30%. This effect of such a low amount of NSAID inthe upregulation of cell survival gene expression is rathercomparable to the effect of proteasome inhibitors likeMG-132or epoxomicin which at nanomolar ranges has been reportedto actually increase the proteasome activity in the pretreatedneocortical neurons [147]. Also, this was the first study thatmeasured COX-2 levels directly in the cardiac cells [146].Although no significant changes in the COX-2 levels wereobserved in cells treated with 1 𝜇M celecoxib, at 10 𝜇M theCOX-2 levels decreased [146].

    Apart from studying the effect of NSAIDs on cardiomy-ocytes, the role of NSAIDs on the COX enzyme system inplatelets is also of prime importance. Platelets are impor-tant for the life-saving blood coagulation process [148] andare closely associated with CVD [149]. The aggregationof platelets by agonists like ADP, collagen, thrombin, orepinephrine leads to the formation of thrombus (microaggre-gate of platelets in fibrin mass), at the site of atheroscleroticplaque fissure on the coronary artery wall of the heart whichresults in thrombosis or CVD [150]. As expected because ofthe importance of platelets in CVD, the generation of ROS inplatelets has been well-studied [149]. Measurement of O

    2

    ∙−

    in platelets activated by thrombin as well as in inactivatedplatelets demonstrated that thrombin significantly increasedthe free radical (O

    2

    ∙−) generation by ∼40% in the activatedplatelets compared to the inactivated platelets [149]. Althoughthis result suggests the possibility of increased ROS formationin activated platelets, no direct role of NSAIDs in theproduction of ROS in the platelets has been reported [151].However, it has been demonstrated that selective COX-2inhibition led to an increase in platelet activation [152]. Theincreased platelet activation by COX-2 inhibition facilitatedthrombotic vessel occlusion after the disruption of the vesselwall supporting a critical role for COX-2 inhibition inplatelet activation and aggregation leading to thrombosis[152]. All these effects were accompanied by a decline in theproduction of endothelial prostacyclin. Diclofenac (1mg/kg),a nonselective NSAID, caused significantly increased plateletvessel wall interaction with an increase in the number ofadherent platelets on the endothelium of diclofenac treatedanimals compared to the control group [153]. Thromboticvessel wall occlusion was also increased in animals treatedwith diclofenac once the vessel wall had been injured [153].Thedirect contribution of ROS induced byNSAIDs to plateletactivation and aggregation is yet to be determined. The roleof aspirin in cardioprotection by the inhibition of plateletaggregation due to the inhibition of platelet thromboxane A2iswell established [154, 155]. Similarly, naproxen has also beensuggested to have significant antiplatelet effect comparable tothat of aspirin [156].

    13. Possible Alternatives to NSAIDs forReduced Cardiovascular Events

    With the availability of a larger selection of synthetic drugs,people are more aware of the side effects of taking drugson a regular basis. Nutraceuticals or phytoceuticals are aclass of compounds that are derived from plants with feweror no side effects compared to synthetic drugs [157] but

  • 18 Oxidative Medicine and Cellular Longevity

    with similar and sometimes even better beneficial biologicaleffects [158]. Hyperforin obtained from the herb Hypericumperforatum has been reported to inhibit COX-1 3–18-foldhigher when compared to aspirin [158]. Hyperforin is anatural antioxidant and has been shown to inhibit excessiveROS generation [159] as well as prostaglandin synthesis [160].Several other studies indicate the usefulness of nutraceuticalsin cardiovascular protection specifically by the inhibition ofCOX. Purified anthocyanins (pigment responsible for thecolor of fruits) have been shown to have an effect on COXactivity [161]. While NSAIDs like naproxen (10 𝜇M) andibuprofen (10 𝜇M) showed COX-1 inhibition of 47.3% and54.3%, respectively, anthocyanins from raspberries (10 𝜇M)and blackberries (10 𝜇M) showed an inhibition of 45.8%and 38.5%, respectively. COX-2 inhibition by naproxen andibuprofen was 39.8% and 41.3%, for sweet cherries it rangedfrom 36% to 48%, and for berries the inhibition was between31% and 46% [161]. The antioxidant activities of the antho-cyanins derived from these fruits were also high suggestingthe importance of their role in reducing ROS.

    In other studies the biologically active compounds ofmycelia of Grifola frondosa and Agrocybe aegerita (ediblemushrooms) were isolated and studied for their antioxidantand COX activity [162, 163]. The authors found that that fattyacids like ergosterol, ergostra-4,6,8(14),22-tetraen-3-one, and1-oleoyl-2-linoleoyl-3-palmitoylglycerol in the case of Grifolafrondosa and palmitic acid, ergosterol, 5,8-epidioxy-ergosta-6,22-dien-3beta-ol, mannitol, and trehalose in the case ofAgrocybe aegerita as determined by spectroscopic analysiswere the most potent in inhibiting both COX-1 and COX-2 [162, 163]. The efficiency of fish oil omega-3 fatty acidslike eicosapentaenoic acid (EPA) and docosahexaenoic acid(DCHA) [164] in preventing CVD, stroke, and high bloodpressure is also well established [165, 166]. It was suggestedthat fish oils have natural inhibitors which can inhibit COX[167]. While the EPA of fish oils inhibited COX-1 more thanCOX-2 [168], DCHA was shown to be an effective inhibitorof arachidonic acid induced prostaglandin biosynthesis [169].Among the other reports available indicating the role ofnutraceuticals in the cyclooxygenase pathway, calcitriol (vita-minD) [170], several types of flavonoids [171, 172], andmarinederived steroids from formosan soft coral Clavularia viridis[173] all have been reported to affect prostaglandin synthesis.Interestingly most of the compounds exhibit antioxidantproperties and inhibit both COX-1 and COX-2 [161, 172]suggesting a role in cardioprotection.

    14. Conclusion

    The review focuses on ROS generated by NSAIDs and its rolein CVD. While the number of clinical experiments investi-gating the effects of NSAIDs on the cardiovascular eventshas significantly increased over the last two decades, basicresearch related to the mechanism by which NSAIDs causecardiovascular dysfunction is limited. High variability inthe clinical trials conducted (different populations, dosages,exposure, and types of NSAIDs) has led to results which aredifficult to interpret and compare between studies. However,irrespective of the type of NSAID used, increased occurrence

    of CVD is common. Clinical trials showed that the risk ofCVD is higher for coxibs than nonselective NSAIDs probablythrough the imbalance between prostacyclin/thromboxanelevels.

    According to the Joint Meeting of the Arthritis AdvisoryCommittee and Drug Safety and Risk Management Advi-sory Committee, US FDA (2014), patients with a historyof myocardial infarction, heart failure, hypertension, andother CV risk factors are at a greater risk of developingcardiovascular diseases due to NSAIDs usage compared tonormal individuals [174]. The committee concluded thatthe increased risk of fatal cardiovascular thrombotic events,myocardial infarction, and stroke by NSAIDS should belessened by using the lowest effective dose of NSAIDs for theshortest period of time possible.

    Interestingly most of the studies that showed the gener-ation of ROS induced by NSAIDs in various types of cellsinvolved nonselective or semiselective NSAIDs (Table 3).However, irrespective of the type of NSAIDs used, eitherselective COX-2 inhibitors or the nonselective NSAIDs,NSAIDs produced oxidative stress with the nonselectiveNSAIDs showing a greater degree of ROS generation [114].Therefore it may be hypothesized that it is through thegeneration of ROS as well as the upregulation and downreg-ulation of several cell survival pathways that these NSAIDsexert their thrombogenic effect. Although COX-2 inhibitorshave been shown to cause CVD some COX-2 inhibitorssuch as celebrex are currently still used widely, as the FDAdetermined the benefits of this drug outweigh the potentialrisks in properly selected and informed patients. The use ofCOX-2 inhibitors underscores the need for compounds withNSAID like properties without the side effects.

    As such, various aspects of NSAID induced cardiotoxi-city still need to be investigated, including the significanceof NSAID induced lipoxygenase ROS generation in thecardiovascular system, determining if prevention of ROSproduction reducesCVDanddetermining if ROSproductionis needed for pain relief. Answers to these questionswill resultin substantial improvement on how CVD risk managementwill be conducted in the future.

    Abbreviations

    CVD: Cardiovascular diseaseCOX-1: Cyclooxygenase-1COX-2: Cyclooxygenase-2NSAIDS: Nonsteroidal anti-inflammatory drugsROS: Reactive oxygen species.

    Conflict of Interests

    The authors declare that there is no conflict of interestsregarding the publication of this paper.

    Acknowledgment

    This research was carried out using UC Davis Researchfunds.

  • Oxidative Medicine and Cellular Longevity 19

    References

    [1] L. J. Crofford, “Use of NSAIDs in treating patients witharthritis,” Arthritis Research & Therapy, vol. 15, supplement 3,article S2, 2013.

    [2] Y. I. Cha and R. N. DuBois, “NSAIDs and cancer prevention:targets downstream of COX-2,”Annual Review of Medicine, vol.58, pp. 239–252, 2007.

    [3] J. Cuzick, F. Otto, J. A. Baron et al., “Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: aninternational consensus statement,” The Lancet Oncology, vol.10, no. 5, pp. 501–507, 2009.

    [4] J. L. Liggett, X. Zhang, T. E. Eling, and S. J. Baek, “Anti-tumor activity of non-steroidal anti-inflammatory drugs:cyclooxygenase-independent targets,” Cancer Letters, vol. 346,no. 2, pp. 217–224, 2014.

    [5] F. L. Lanza, F. K. L. Chan, and E. M. M. Quigley, “Guidelinesfor prevention of NSAID-related ulcer complications,” TheAmerican Journal of Gastroenterology, vol. 104, no. 3, pp. 728–738, 2009.

    [6] &𝑎𝑝𝑜𝑠; 𝐴. Lanas, P. Carrera-Lasfuentes, Y. Arguedas et al., “Riskof upper and lower gastrointestinal bleeding in patients takingnonsteroidal anti-inflammatory drugs, antiplatelet agents, oranticoagulant,” Clinical Gastroenterology and Hepatology, 2014.

    [7] F. Lapi, L. Azoulay, H. Yin, S. J. Nessim, and S. Suissa, “Concur-rent use of diuretics, angiotensin converting enzyme inhibitors,and angiotensin receptor blockers with non-steroidal anti-inflammatory drugs and risk of acute kidney injury: nestedcase-control study,” British Medical Journal, vol. 346, no. 7890,Article ID e8525, 2013.

    [8] K. A. Taubert, “Cardiology patient pages: can patients with car-diovascular disease take nonsteroidal antiinflammatory drugs?”Circulation, vol. 117, no. 17, pp. e322–e324, 2008.

    [9] M. Soyun, J. E. G. Hwang, Z. Cui, and A. V. Gomes, Non-Steroidal Anti-Inflammatory Drugs and Increased Risk of SuddenCardiac Death, Nova Science Publisher, Hauppauge, NY, USA,2013.

    [10] F. E. Silverstein, G. Faich, J. L. Goldstein et al., “Gastrointestinaltoxicity with celecoxib vs nonsteroidal anti-inflammatory drugsfor osteoarthritis and rheumatoid arthritis: the CLASS study:a randomized controlled trial,” The Journal of the AmericanMedical Association, vol. 284, no. 10, pp. 1247–1255, 2000.

    [11] L. A. G. Rodr&𝑎𝑝𝑜𝑠; 𝑖guez and L. B. Tolosa, “Risk of upper gas-trointestinal complications among users of traditional NSAIDsand COXIBs in the general population,” Gastroenterology, vol.132, no. 2, pp. 498–506, 2007.

    [12] J. L. Goldstein, F. E. Silverstein, N. M. Agrawal et al., “Reducedrisk of upper gastrointestinal ulcer complications with cele-coxib, a novel COX-2 inhibitor,” The American Journal ofGastroenterology, vol. 95, no. 7, pp. 1681–1690, 2000.

    [13] A. Helin-Salmivaara, A. Virtanen, R. Vesalainen et al., “NSAIDuse and the risk of hospitalization for firstmyocardial infarctionin the general population: a nationwide case-control study fromFinland,” European Heart Journal, vol. 27, no. 14, pp. 1657–1663,2006.

    [14] M. Amer, V. R. Bead, J. Bathon, R. S. Blumenthal,