oxidative stress and inflammation in osa.pdf

Upload: cristianamihaila

Post on 04-Jun-2018

226 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    1/21

    Chapter 21

    Oxidative stress and

    inflammation in OSAL. Lavie

    Summary

    Enormous progress has been made in the last decade inunderstanding the impact obstructive sleep apnoea syndrome

    (OSAS) has on the cardiovascular system and the associatedcomorbidities. However, the mechanisms governing theseassociations are poorly understood. The accumulated evidenceimplicates oxidative stress and inflammation as two basicmechanisms associated with OSAS and with various metabolicdysregulations. The recent development of various experimen-tal models of intermittent hypoxia, based on animals andcultured cells, has helped to unveil some of these mechanismsand pathways affected by the intermittent hypoxia. It is likelythat an improved understanding of these mechanisms may leadto the development of new and more effective treatmentmodalities to abort the cardiovascular risk, resulting fromoxidative stress and inflammation.

    Keywords: Adhesion molecules, cardiovascular morbidity,cytokines, inflammation, obstructive sleep apnoea, oxidativestress

    Correspondence: L. Lavie, Unit ofAnatomy and Cell Biology. The Ruthand Bruce Rappaport Faculty ofMedicine, Technion, POB 9649,

    31096, Haifa, Israel,Email [email protected]

    Eur Respir Mon 2010. 50, 360380.Printed in UK all rights reserved.Copyright ERS 2010.European Respiratory Monograph;ISSN: 1025-448x.DOI: 10.1183/1025448x.00025509

    The increased awareness of the impact obstructive sleep apnoea syndrome (OSAS) has on thecardiovascular system, quality of life, and its association with risk factors such as obesity andhypertension has lead to intensive research on these associations and the mechanisms involved [1].OSAS, a sleep-breathing disorder, is characterised by intermittent and recurrent pauses inrespiration. Based on this trait that implicates the apnoea-related multiple cycles of hypoxia/reoxygenation with an increased production of reactive oxygen species (ROS), similar toischaemia/reperfusion, it was proposed that oxidative stress and inflammation play a key role [2].However, oxidative stress and inflammation are also fundamental to the development ofatherosclerosis, cardiovascular and other morbidities associated with the metabolic syndrome.This oxidative/inflammatory cycle, which leads to the development of atherosclerosis andcardiovascular morbidity in OSAS due to the intermittent hypoxia (IH), is illustrated in figure 1.

    This chapter covers the main findings in this area of oxidative stress/inflammation and itsconsequences in OSAS. These may lead to the development of endothelial dysfunction and earlyclinical signs of atherosclerosis and eventually, cardiovascular morbidity and mortality. The potentialrole of various inflammatory blood cells in initiating and propagating oxidative stress/inflammation

    360

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    2/21

    is also described and a discussionis devoted to the potential role ofassociated comorbidities in exacer-bating these effects.

    Obstructive sleep

    apnoea andcardiovascularmorbidity

    In recent years OSAS has emerged asa major public health problem, dueto its profound impact on patientshealth and quality of life [3, 4].OSAS is primarily characterised byrecurrent pauses in respiration,

    which result in cyclic decreases inblood oxygen saturation that isdenoted as IH, and brief arousalsfrom sleep due to obstruction of theupper airway. The severity of OSASis defined by the apnoea/hypopnoeaindex (AHI), which denotes thenumber of apnoeas plus hypop-noeas divided by hours of sleep.The prevalence is particularly high

    in males, but is also high due tocentral obesity, smoking and inpost-menopausal females. It is esti-mated that 4% of males and 2% offemales have at least five disordered-breathing events in each hour ofsleep in the form of apnoeas or hypopnoeas, accompanied mostly by day time somnolence [3, 5]. Butthe prevalence may be higher in non-symptomatic subjects, increasing to 24% and 9% of adult malesand females, respectively [3, 5]. In the obese and the elderly these values may increase to 60% [36].

    Notably, OSAS has profound effects on the cardio-cerebrovascular system. This is based on a greatnumber of small and large scale cross-sectional, prospective, population-based and interventionstudies, which identified OSAS as an independent risk factor for cardiovascular morbidity andmortality [1]. Generally, OSAS is closely associated with an increased risk for hypertension,ischaemic heart disease, strokes, arrhythmias, chronic heart failure, as well as cardiovascularmortality. However, the impact of OSAS on the cardiovascular system is mainly evident inrelatively young populations, i.e. aged 50-yrs-old or younger, and less so with the elderlypopulation [7]. Moreover, evidence from recent years has also shown that OSAS is associated withadditional risk factors of the metabolic syndrome, such as hyperlipidemia, insulin resistance,hypertension and obesity, which also promote cardiovascular morbidity. These additional riskfactors, when clustered with OSAS, may further augment the risk for cardiovascular morbidity byacting synergistically with the apnoeic events [810].

    Since the development of cardiovascular morbidity is of a multifactorial nature it could be largelyaffected by the genetic makeup, associated comorbidities and lifestyle related variables, such as physicalactivity, smoking and diet [11]. However, based on the ramifications of the IH, which is the hallmark ofOSAS, cardiovascular morbidity could be the outcome of such hypoxic events, although additional

    OSAS/intermittent hypoxia

    ROS

    Endothelial dysfunction

    Atherosclerosis

    Activation

    Adhesion

    Injury

    Cardiovascular morbidity

    Cytokines, adhesion molecules

    Transcription factor activation: NF-BAP-1

    Leukocytes,platelets

    Endothelialcells

    Inflamm

    ation

    Figure 1. Oxidative stress and inflammation in obstructive sleepapnoea syndrome (OSAS). The intermittent hypoxia inducesreactive oxygen species (ROS) formation, which in turn activate an

    inflammatory cascade via activation of transcription factors and

    downstream genes as inflammatory cytokines and adhesionmolecules. These in turn can further activate transcription factors

    and various blood cells. Activated leukocytes and platelets producehigher amounts of ROS, adhesion molecules and pro-inflammatory

    cytokines, exacerbating this oxidative-inflammatory cycle andfacilitating endothelial dysfunction, which is the prelude to athero-

    sclerosis and cardiovascular morbidity [2]. NF-kB: nuclear factor-kB;AP-1: activator protein 1.

    361

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    3/21

    factors could also contribute. Early studies have established that OSAS is accompanied by activation ofthe sympathetic nervous system, swings in intrathoracic pressure and altered blood coagulability [12].Indeed these measures were all shown to increase with the severity of OSAS and could largely contributeto cardiovascular morbidity. Owing to the fact that treatment with nasal continuous positive airwaypressure (nCPAP) attenuated some of these measures, it clearly attests to their significance [13, 14].Although these mechanisms contribute to the development of endothelial dysfunction, atherosclerosisand cardiovascular morbidity in OSAS, a large body of evidence supports a role for oxidative stress and

    inflammation as the initiating factors. This is mainly attributed to the fact that oxidative stress, which ispromoted in conditions such as ischaemia/reperfusion, is also manifested in OSAS due to the IH. Onceoxidative stress ensues it elicits inflammation and vice versa, inflammation promotes oxidative stress,thereby creating a vicious cycle by exacerbating each other. Additionally, both oxidative stress andinflammation are critical components in eliciting endothelial dysfunction and promotingatherosclerosis and consequently cardiovascular morbidity, as described in figure 1.

    ROS and oxidative stress in physiology/pathphysiology

    ROS molecules and oxidative stress have a central role in normal cellular function, as well as in various

    pathological conditions. In the last two decades their key role as signalling molecules, which activateand control various transduction pathways in physiological as well as pathophysiological conditions,has been increasingly established. However, it should be acknowledged that this is still a controversialarea and while severe oxidative stress can certainly activate these pathways it is not entirely clear if ROSplay a role in the physiological signalling processes, most likely due to the large intracellular pool ofantioxidants. Yet in pathological conditions the excessive formation of ROS is prone to damagingsurrounding tissues by promoting signalling and activation of inflammatory pathways [15].

    Sources and activity of ROS

    Free radicals or ROS are atoms or molecules with one or more unpaired electron in their outerorbit. This trait enables them to chemically react with other molecules [15]. When two freeradicals react with each other a nonradical molecule is formed, but when a radical reacts with anonradical molecule the product is a new radical that can react with either another radical or anonradical, thereby propagating free radical chain reactions.

    The most abundant ROS molecule is the superoxide (O2N-) radical. It is formed by a univalent

    reduction of molecular oxygen, mainly during normal aerobic metabolism. Thus, mitochondrialrespiration is its major source. It is estimated that,5% of cellular ROS molecules are by-products ofnormal mitochondrial respiration. However, additional enzymatic systems also produce superoxide.

    NADPH oxidase from primed leukocytes and endothelial cells is another major source ofsuperoxide [2, 16]. Other enzymes include xanthine oxidase and possibly uncoupled endothelialnitric oxide synthase (eNOS) [2, 16]. Although the superoxide is relatively a weak radical, itsreaction with other molecules yields additional, more potent ROS molecules and oxidants, such ashydrogen peroxide (H2O2), hydroxyl radical (OH

    ?) and lipid peroxides, when reacting with lipids.Apart from ROS, also nitrogen reactive species (RNS) are considered free radicals and they toocontribute to oxidative-nitrosative stress. One such toxic molecule with relevance to endothelialfunction is peroxynitrite (OONO-), which is formed by the reaction of the potent vasodilator nitricoxide (NO) with superoxide. Consequently, the availability of NO is diminished and properendothelial function is hampered [17]. A schematic and simplified illustration describing the

    possible sources for ROS/RNS production and their fate in OSAS is presented in figure 2.

    Oxidative stress

    As stated previously, the formation of ROS is as a by-product of normal cellular respiration andaerobic metabolism. Thus, various antioxidant defense mechanisms have evolved to counteract

    362

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    4/21

    excessive formation of ROS andsustain the oxidation-reduction(redox) status of a cell under strictregulation.

    Antioxidants can be divided intoenzymes, such as superoxide dismu-tase (SOD), catalase and glutathione

    peroxidase, and small molecules,e.g.vitamins C and E, glutathione anduric acid. Together these help pre-serve the tightly regulated balancebetween oxidants and antioxidants.Thus, oxidative stress is viewed as adisruption of this balance throughexcessive production of ROS. Suchoverproduction of ROS/RNS mayhinder cell activity and function

    by damaging various biomolecules,such as DNA, lipids, proteins andcarbohydrates [15].

    The signallingfunctions of ROS

    ROS act like a double-edged sword,apart from injury to macromole-

    cules and cellular components, ROSare vital regulators of a plethora ofsignal-transduction pathways andfunction as second messengers inmany physiological, as well as path-ological, conditions [15, 18]. More-over, their importance in maintaining stringent cellular redox homeostasis is well established[15, 19]. ROS were implicated in the activation signalling pathways involved in the initiation ofinflammatory and adaptive pathways, through activation of various nuclear transcription factors,such as hypoxia inducible factor (HIF)-1a, nuclear factor (NF)-kB, activator protein (AP)-1, sterol

    regulator element binding proteins (SREBPs), GATA-4 [15, 2022] and NF-erythroid derived 2-related factor (Nrf2)-antioxidant responsive element (ARE), that regulates antioxidant genes [23].These pathways are critical to the development of atherosclerosis in many pathological conditions,including those of the metabolic syndrome [24]. Additional information on transcription factorsinvolved with ROS molecules, with potential importance to OSAS pathology, have been describedpreviously [25]. However, it should be stressed that activation of these transcription factors is notsolely dependent on ROS and in some instances there is a controversy regarding the function of ROSas activators, as is specified later. Nonetheless, the interactions of transcription factors with ROS areintricate and intermingled [26].

    Effects of hypoxia/reoxygenation and intermittent hypoxiaIschaemia/reperfusion or hypoxia/reoxygenation is one of the most fundamental and relevantdiscoveries to OSAS. It established the involvement of superoxide radicals in injury of variousmacromolecules and cellular components in conditions that involve ischaemia or hypoxia followedby reperfusion/reoxygenation due to over production of ROS and subsequently oxidative stress [27].

    ++ +

    SOD

    O2-

    Lipids

    ONOO-

    +

    Cat/GPx

    OSAS/intermittent hypoxia

    Mitochondrialdysfunction

    Xanthineoxidase

    NADPHoxidase

    Lipidperoxidation

    NO

    H2O2

    H2O+O2

    Figure 2. A schematic illustration of reactive oxygen species(ROS) and nitrogen reactive species production in obstructive sleep

    apnoea syndrome (OSAS), during intermittent hypoxia, describingthe main sources of ROS. The superoxide (O2

    N-) formed can react

    with lipids to induce lipid peroxidation and a reaction with nitric

    oxide (NO) promotes the formation of peroxynitrite (ONOO-).

    Superoxide dismutase (SOD) sequestrates the superoxide yieldinghydrogen peroxide (H2O2), while catalase (Cat) and glutathione

    peroxidase (GPx) may convert the hydrogen peroxide to H2O and

    O2. Note the equations are not balanced. NADPH: reducednicotinamide adenine dinucleotide phosphate.

    363

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    5/21

    Primarily, the relevance of ischaemia/reperfusion injury was shown in conditions such asischaemic heart disease, stroke, surgery, and organ transplantation. In the past 20 yrs at least 80pathologies were associated with excess ROS and oxidative stress. ROS were shown to increase inall inflammatory diseases studied, in ischaemic heart diseases, stroke, cancers, diabetes,hypertension, neurodegenerative disorders and atherosclerosis [28]. In the last decade, OSAS hasalso been shown to contain an oxidative stress component [22]. It was suggested that theseintermittent breathing arrests during sleep, which are accompanied by drastic changes in arterial

    oxygen tension, bear similarities to ischaemia/reperfusion processes and may therefore promotethe formation of ROS, thereby inducing oxidative stress [2]. Consequently, the critical role ofROS molecules in inducing inflammatory pathways through increased activation of variousinflammatory blood cells and endothelial cells in OSAS was investigated. Such sequelae weresuggested to lead to the development of endothelial dysfunction and thereby enhance cerebro/cardiovascular morbidities [29].

    Oxidative stress in OSAS

    Sources and targets of ROS in IH and OSAS

    There is ample evidence that ROS molecules are increased in response to IH and in OSAS. Studiesin cells in culture, organs, animal models that mimic OSAS and in humans all demonstrated thatOSAS and IH can induce mitochondrial dysfunction and thereby increase oxidative stress [3034].In many animal models NADPH oxidase was also reported to increase in tissues like the brain andcarotid body [3537]. In humans there is indirect evidence for NADPH oxidase activation invarious inflammatory cells [3840]. There is also indirect evidence implicating activation ofxanthine oxidase in OSAS by using allopurinol, which is a specific xanthine oxidase inhibitor [41],and identifing some of its metabolic by products in the plasma [2, 42].

    The primary targets of oxidative stress in OSAS are likely to be endothelial cells and the

    vasculature in general. However, since IH also induces inflammatory cell activation, activatedleukocytes could in turn contribute to increased oxidative stress in the vasculature. Yet, studies inanimal models utilising IH have shown that in tissues, such as the heart, brain and the liver,oxidative stress is also increased [37, 4347]. But since mitochondrial dysfunction is a likely sourceof ROS in IH/OSAS, it is reasonable to assume that each cell type would eventually be affected,depending on the severity of OSAS and the individual sensitivity of each cell type or organ, to thedecreased oxygen levels and ROS generation [45].

    Evidence for ROS and oxidative stress

    As aforementioned, the production of ROS and oxidative stress in OSAS are attributed to the cyclicnature of the multiple intermittent hypoxia episodes that resemble ischaemia/repefusion. Indeed a greatnumber of studies in the last decade clearly point to increased ROS formation and oxidative stress as aresult of this trait of OSAS. However, only a limited number of studies have shown a direct increase inROS formation by OSAS patients by utilising inflammatory cells, which primarily function in hostdefense and release large quantities of ROS [3840]. A study by SCHULZ et al. [40] demonstratedincreased ROS production in OSAS stimulated neutrophils that was attenuated after nCPAP treatment.In the studies conducted by DYUGOVSKAYA and co-workers [38, 39], stimulated monocytes andneutrophils and nonstimulated monocytes of patients with OSAS released higher amounts of ROS ascompared with controls. However, most of the data regarding increased ROS and oxidative stress inpatients with OSAS is provided by indirect evidence; mainly from circulating markers of oxidative stress.

    Lipid peroxidation is a sensitive marker due to the high liklihood of lipids to undergo oxidation,and therefore is a highly used oxidative stress marker. Thus, increased oxidative stress in OSAS wasprimarily shown by using various markers of lipid peroxidation in plasma and serum [4850].Also, the levels of circulating oxidised low-density lipoprotein (oxLDL) were shown to increase in

    364

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    6/21

    patients with OSAS [51, 52]. Oxidative stress was also demonstrated in DNA by an increasedurinary excretion of 8-hydroxy-29deoxyguanosine [53] and in exhaled breath [54]. Treatment withnCPAP or a dental device attenuated the oxidative stress markers [48, 49, 54, 55]. Increasedoxidative stress and lipid peroxidation in OSAS was also associated with an increase in carotidintima media thickness [56]. Moreover, in young healthy adults exposed to 4 days of IH,overproduction of ROS modulated increased acute-hypoxic ventilatory responses [57]. Alsogenomic microarray studies indicate that oxidative stress was increased in OSAS [58].

    Diminished antioxidant capacities, which disrupt the tightly regulated balance and promoteoxidative stress, were also described in OSAS. Total antioxidant capacity in OSAS was reported todecrease in a number of studies and lowered levels of vitamins A and E were also described [50, 59].Additionally, the ability of serum albumin to act as an antioxidant was impaired in OSAS and wasimproved after nCPAP treatment [60]. Also, the activity of the antioxidant enzyme paraoxonse(PON)-1 was attenuated in OSAS [48]. This finding is in line with the increases observed in oxLDLlevels and the appearance of dysfunctional high-density lipoprotein (HDL) in OSAS [51]. SincePON-1 is physically bound to HDL and protects both LDL and HDL from oxidative modification.Importantly, PON-1 activity was also significantly correlated negatively with the severity of thesyndrome but not with age or body mass index (data not shown). Although in a small number of

    studies negative results were reported, the overwhelming number of studies presenting positivefindings clearly attest to altered ROS balance and increased oxidative stress in OSAS.

    Oxidative stress and endothelial dysfunction

    NO is a potent vasodilator that regulates and maintains vasodilatation of blood vessels andattenuates expression of adhesion molecules. These properties prevent vasoconstriction andinhibit leukocytes/endothelial cells interactions. Thus, NO maintains a healthy and functionalendothelium. Attenuated NO levels in OSAS were reported in several instances [6163]. Thesestudies indicate that NO bioavailability is compromised, most likely through deactivation by ROS,

    which result in endothelial dysfunction [2, 64]. This assumption was reinforced by a more recentstudy demonstrating that the activity of eNOS and its phosphorylated active form (p-eNOS) wereattenuated in freshly harvested venous endothelial cells from patients with OSAS. Moreover, theoxidative stress marker nitrotyrosine, which is indicative of NO inactivation by superoxide wasalso elevated [65]. More recently these findings were expanded to show that untreated OSAS,rather than obesity, was a major determinant of vascular endothelial dysfunction and increasedoxidative stress [66]. It is noteworthy that in both studies treatment with nCPAP improvedendothelial function, as determined by flow mediated dilation of the brachial artery [65, 66].Accordingly, treatment of OSAS patients with antioxidants, such as the xanthine oxidase inhibitorallopurinol, or by injecting a single bolus of vitamin C, improved endothelial function [41, 67].

    Also, treatment with nCPAP was shown to reverse the diminished serum nitrite and nitrate inobese patients with OSAS [68]. These findings are in line with the involvement of oxidative stressin inducing endothelial dysfunction in OSAS.

    Furthermore, both endothelial dysfunction, which is the subclinical condition that precedesatherosclerosis, and atherosclerosis were reported in patients with OSAS [6972]. For instance,measures, such as increased intima-media thickness, arterial plaque formation and calcified arteryatheromas, were documented in OSAS patients free of comorbidities or cardiovascular disease [7376].Treatments with nCPAP or a dental device improved some of these subclinical signs [55, 77, 78].

    Inflammatory and adaptive pathways in OSASNF-kB

    NF-kB is a pleiotropic transcription factor and a major regulator of a great number of vital cellularfunctions such as proliferation, death and immune responses. It has a key role in initiating

    365

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    7/21

    inflammation by inducing expression of adhesion molecules, inflammatory cytokines andadipokines, which promote atherosclerotic sequelae [79]. NF-kB is activated by a diverse array ofstimuli including inflammatory cytokines and tumour necrosis factor (TNF)-a in particular aswell as various adhesion molecules and ROS. However, in many instances the participation of ROSremains a controversial issue, in particular when studying various cell types, e.g.cancer cells [80].Yet, ROS-dependent NF-kB activation was recently described in human endothelial cells derivedfrom obese individuals [81]. Thus, it appears that ROS dependent NF-kB activation is cell type-

    specific and cell state-specific.In patients with OSAS the upregulated expression of NF-kB was demonstrated in neutrophils andmonocytes [8284]. More recently increased expression of NF-kB was detected in venousendothelial cells obtained from obese patients with OSAS that were compared with obese non-OSAS subjects, demonstrating the contribution of OSAS, rather than obesity, to its upregulation[66]. Additionally these finding were verified in neutrophils of healthy subjects that were treatedwith IH in vitro, specifically implicating IH in activating NF-kB, rather than other causes orproposed mechanisms such as sleep fragmentation or sympathetic activity [85]. In addition,increased expression of adhesion molecules and inflammatory cytokines and adipocytokines, thegene products of NF-kB, was also noted in OSAS [38, 86, 87]. Such increases in the gene productsof NF-kB clearly indicates that activation of NF-kB is associated with OSAS [38, 39, 88, 89]. It isalso noteworthy that a study by RYANet al.[90], who used HeLa cells that were treated with IH invitro, demonstrated a selective activation of NF-kB over HIF-1a, directly implicating IH withupregulated NF-kB expression [90].

    HIF-1a

    The transcription factor HIF-1a is the master regulator for cellular oxygen homeostasis andadaptive responses to hypoxia. Its activity is upregulated and stabilised under hypoxic conditions,

    while regulating hundreds of genes that facilitate oxygen supply to ischaemic/hypoxic tissues [91].Under IH conditions its activation was mainly shown in cells in tissue culture and in experimentalmodels of rodents shown in reviews by SEMENZA and PRABHAKAR[92] and PRABHAKARet al. [93].However, these findings were inconsistent [90, 9496]. Obviously, using various cell types andapplying different patterns and intervals with more or less extreme exposure to IH for varyingdurations of hypoxia and reoxygenation may lead to such inconsistencies [90].

    Unlike in tissue culture, studies conducted on animal models treated with chronic IH reveal acritical role for HIF-1a. In the myocardium, HIF-1aactivity and endothelin-1, a gene product ofHIF-1a, were increased in spontaneously hypertensive rats exposed to chronic IH [97]. Of note,HIF-1a was implicated in hypertension [98] and in components of the metabolic syndrome inrodents treated with chronic IH [99]. However, studies demonstrating activation of HIF-1a inpatients with OSAS were not described thus far. Yet, upregulated expression of some of its geneproducts attests to a possible involvement of HIF-1a. However, again regarding some of the HIF-1agene products investigated in OSAS, such as erythropoietin and vascular endothelial growth factor(VEGF), data are not uniform in all studies [2, 100102].

    Nuclear factor erythroid derived 2-related factor

    Another transcription factor with potential implications to OSAS is Nrf2. This is a true redox

    regulated transcription factor that is ubiquitously expressed at low levels in various cells andtissues. It regulates a great number of antioxidant genes and drug-detoxifying enzymes, and bythat serves a cellular protective mechanism. Importantly, it is also activated in response toenvironmental and endogenous stresses and thus helps to maintain health by preventing varioushuman diseases, such as cardiovascular, neurodegenerative, cancer as well as inflammatory andischaemic conditions [103].

    366

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    8/21

    Two recent studies in tissue culture implicated upregulated expression of Nrf2 under IHconditions [96, 104]. Although the intervals of hypoxia and reoxygenation employed did notmimic IH as in OSAS, nonetheless the findings described are interesting. In the first study usinghuman aortic endothelial cells, gene expression profiles were determined. By using IH cyclesalternating between 10 min hypoxia and 10 min reoxygenation Nrf2 gene expression wasupregulated by more than three-fold over normoxia but classical genes regulated by HIF-1 werenot [96]. In the second study, which was conducted on human lung adenocarcinoma A549 cells by

    using 2-h periods and alternating between hypoxia and reoxygenation, Nrf2 as well as HIF-1 werefound to be upregulated. Moreover, NADPH oxidase derived ROS were implicated in theupregulation of Nrf2 [104]. These studies clearly emphasise the relevance of the different patterns,durations and the severity of the IH employed as determinants for activation of distinct pathways,such that may lead to contradictory findings in OSAS.

    It is noteworthy that there is a crosstalk between the various transcription factors mentionedabove. For instance, while hypoxia upregulates the transcription of HIF-1a by an NF-kBdependent mechanism, also the NF-kB pathway is affected by the HIF pathway [105, 106]. Suchfindings may also implicate the participation of HIF-1a in inflammatory pathways [25]. Also,there appears to be a crosstalk between Nrf2 and NF-kB, since the Nrf2 and NF-kB signalling

    pathways interface at several points to control the transcription or function of the downstreamtarget proteins [103]. Also, thioredoxin-1, the downstream gene of Nrf2, has been shown to inducethe amount of HIF-1a protein and thereby may indicate a possible crosstalk between Nrf2 andHIF-1a[107]. These complexities in the interactions between the various transcription factors canillustrate the difficulties in teasing out the function of each pathway under IH conditions. Findingsdescribing some of the possible intricate interactions between these transcription factors have beendescribed elsewhere [103, 108].

    Activation of cellular inflammatory pathways in OSAS

    As stated above, increased expression of ROS and augmented oxidative stress activate a plethora ofinflammatory pathways. Notably, NF-kB activation in OSAS is a prominent feature that promotesover-expression of adhesion molecules. These facilitate recruitment and accumulation of bloodcells on the endothelium lining the vasculature. Activation of NF-kB also promotes increasedexpression of pro-inflammatory cytokines that further exacerbate endothelial cells/blood cellsinteractions leading to endothelial cell injury and dysfunction [2].

    Typically, circulating blood cells express low levels of adhesion molecules and intracellularinflammatory cytokines enabling blood cells to flow freely in the circulation, while withstandinginteractions with endothelial cells. However, expression of adhesion molecules and cytokines is

    upregulated by a great number of stimuli. Infections, cytokines, hypoxia/reoxygenation, as well assleep apnoea were shown to increase the expression of various adhesion molecules and otherinflammatory molecules in blood cells and endothelial cells.

    Adhesion molecules and blood leukocytes/endothelial cellinteractions

    The expression of adhesion molecules is a highly regulated and sequential process. It facilitatesinteractions between activated blood cells and endothelial cells, promoting adhesion and injury to

    the vascular endothelium. The most notable groups of adhesion molecules include the selectins.There are three major groups these are: 1) the L-selectins that are expressed on leukocytes, 2) the E-selectins in endothelial cells, and 3) P-selectins that are expressed in platelets as well as endothelialcells. This group of adhesion molecules promotes the initial interactions allowing for weak bindingbetween leukocytes and endothelial cells. However, the firm binding to endothelial cells is mediatedby the integrins, which also enables transmigration into the interstitial layer through the endothelial

    367

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    9/21

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    10/21

    fundamental injury-limiting mechanism to control inflammation, its suppression together withincreased expression of selectins may heighten PMNs/endothelial cells interactions in OSAS, andaugment their destructive potential towards the endothelium [39].

    Monocytes

    Monocytes also participate in host defense mechanisms by acting as professional phagocytes. However,

    they also participate in various stages of atherosclerosis and contribute to its development throughactivation and foam cell formation [24, 113]. Similar to PMNs, also monocytes of patients with OSASdisplay activation and increased expression of selectins (CD15), integrins (CD11c), ROS, cytokines andincreased adhesion to endothelial cellsin vitro[38, 89, 114], while nCPAP treatment effectively lowersadhesion molecules expression (CD15, CD11c) and adhesion to endothelial cells (table 1) [38].

    Cytotoxic T-lymphocytes

    Several subpopulations of T-lymphocytes were shown to be prevalent in atherosclerotic lesionsand to modulate atherosclerotic responses [115, 116]. These were mainly implicated in variousatherogenic processes via cytokine secretion and antibody production. Table 1 presents asummary of the findings on CD8+, CD4+, andcdT-cells of patients with OSAS. In fact, all T-cellsinvestigated expressed an activated and a cytotoxic phenotype in OSAS [88, 110, 111]. However,OSAS cd T-cells expressed the highest cytotoxic potential against endothelial cells as comparedwith OSAS CD8+, and the cytotoxicity of CD4+ T-cells was the lowest.

    Platelets

    Similarly to white blood cells, platelets were shown to be activated in OSAS. In physiologicalconditions platelets are circulating in a quiescent state protected from activation by inhibitorymediators released from intact endothelum. This helps to maintain vascular homeostasis.

    However, platelets rapidly undergo activation due to oxidative stress, endothelial dysfunction orvascular damage. Subsequently, increasing the interactions with monocytes and PMNs is followedby increased adhesion and aggregation at the vessel wall [117]. In OSAS patients, plateletsexpressed increased activation, adhesion molecules and aggregability in vitro. The most notableadhesion molecule, which is upregulated in activated platelets of OSAS, is the P-selectin (CD62P)that was attenuated by nCPAP treatment [118121]. Additional measures of atherosclerosis, suchas increased blood viscosity, fibrinogen and hematocrit, were also heightened. These may furthercontribute to the increased incidence of cardiovascular events in OSAS [122124].

    Endothelial cells

    The functional endothelium consists of a cell layer that protects the vasculature by providing apermeability barrier. It regulates vessel tone and maintains an anti-inflammatory and antithromboticphenotype. While in their nonactivated state endothelial cells resist adhesion to various blood cells.However, activation or injury by various factors including hypoxia/reoxygenation, triggers theexpression of adhesion molecules [125, 126]. A number of adhesion molecules and inflammatorymarkers derived from endothelial cells were identified in the circulation of patients with OSAS. Theseincluded E-and P-selectin [127, 128], intracellular adhesion molecule-1 (ICAM-1), and vascular celladhesion molecule-1 (VCAM-1) [127, 129131]. Such circulating adhesion molecules were identifiedas markers of active atherosclerotic diseases, and as predictors of future cardiovascular events [64]. Also

    in activated endothelial cells that were directly harvested form the vasculature of OSAS patients,increased oxidative stress and inflammation were noted. Moreover, nitric oxide bioavailability andtheir repair capacity were compromised [65]. More recently several studies addressed the presence ofendothelial progenitor cells (EPCs) and their repair capacity in the circulation of OSAS patients.However, these studies did not yield conclusive findings, most likely due to the very low numbers inthe circulation [132134]. Interestingly, in a recent study conducted on children with OSAS two

    369

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    11/21

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    12/21

    Inflammatory cytokines and adipokines in OSAS

    Inflammatory cytokines

    Expression of pro-inflammatory cytokines, like adhesion molecules, is also affected by oxidativestress resulting in activation of NF-kB in various blood cells and endothelial cells. While adhesionmolecules facilitate the recruitment and accumulation of platelets and leukocytes to injure the

    endothelium, cytokines may further augment this damage to endothelial cells.

    The cytokines are multipurpose molecules. They are synthesised and released by many cell typesand regulate both the innate and adaptive immune system by complex interactions with varioustranscription factors and cytokines. Many of their functions including macrophage activation,smooth muscle cell proliferation, diminished nitric oxide activity and endothelial cell activationare involved in the progression of atherosclerosis.

    TNF-a is one of the most important cytokines and has a key role in a plethora of cellularfunctions. It is synthesised by a variety of cells including inflammatory leukocytes and adipocytesand is involved with the initiation and progression of cardiovascular pathology [137, 138].

    Additionally TNF-ais capable of inducing oxidative stress, expression of adhesion molecules andstimulating cytokine production viaNF-kB dependent pathways.

    In OSAS, circulating TNF-a levels in serum or plasma were increased [86, 139, 140]. Also, thelevels of other inflammatory cytokines, such as interleukin (IL)-6 and IL-8 were increased.Conversely, the levels of the anti-inflammatory cytokine IL-10 were attenuated in OSAS [73, 87,140142]. Since IH and OSAS initiate the inflammatory response, these pro-inflammatorycytokines can in turn activate NF-kB thereby further augmenting inflammation (fig. 1).

    Apart from circulating levels of cytokines, elevated cytokines levels were described in monocytes andin various cytotoxic T-lymphocytes [110, 111, 114]. Increased TNF-aand IL-8 and decreased IL-10

    levels were detected incd+ T-lymphocytes of patients with OSAS [111]. TNF-awas also increased inCD8+T-cells but did not change in CD4+T-cells [88, 143]. Thus, when using specific cell sourcesundoubtedly TNF-a and other pro-inflammatory cytokines increase while anti-inflammatorycytokines decrease. Still each cell type has a unique profile of cytokine expression [87].

    Inflammatory cytokines/adipokines and obesity

    Cytokines released from adipose tissue are termed adipocytokines or adipokines. The mainadipokines include TNF-a, IL-6, CRP, adiponectin, leptin, resistin, and angiotensinogen [144].However not all adipokines are synthesised exclusively by adipose tissue. Most circulating

    adipokines levels were shown to increase in obese patients and in patients with OSAS, whileanti-inflammatory adipokines such as adiponectin decreased resulting in low-grade inflammationin both instances [87, 145]. As, apart from leukocytes, the adipose tissue is a major source forcytokines/adipokines, determination of circulating levels in OSAS does not allow the identificationof their source. Moreover, OSAS patients are mostly obese individuals. Therefore, it is not clearwhether adipokines are synthesised due to obesityper seand/or because of OSAS. Given that inmost studies cytokines/adipokines levels were determined in the circulation of OSAS patients, thevalues obtained reflect the overall pool of cytokines/adipokines released from variousinflammatory cells, adipocytes, the liver, and other tissues. Obviously such findings do notprovide information on specific inflammatory/anti-inflammatory responses, or on an ongoing

    process in a specific inflammatory cell [87].

    Inflammatory adipokines in OSAS

    One of the most studied adipokines in OSAS is adiponectin. It is an anti-inflammatory andantidiabetogenic cytokine with cardiovascular protective effects. It is the most abundant of all

    371

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    13/21

    circulating adipokines, and is exclusively produced by the adipose tissue. Adiponectin wasshown to decrease in obese individuals and in patients with type 2 diabetes [146, 147]. Due toits anti-inflammarory properties, adiponectin also attenuates the levels of adhesion moleculesand pro-inflammatory cytokines as TNF-a, IL-8 and IL-6. In parallel it increases the levels ofthe anti-inflammatory cytokine IL-10.

    In OSAS, adipokines levels were mainly unaffected by the syndrome. This is particularly evident instudies selecting nonobese, comorbidity free patients [39, 148, 149]. Notably, decreased

    adiponectin levels in OSAS were attributed to obesity [149]. Thus, long-term treatment withnCPAP did not affect adiponectin levels [150]. However in obese patients with OSAS treatmentwith nCPAP reversed the diminished plasma adiponectin levels [68]. Collectively these findingssuggest that obesity, rather then OSAS, is the determining factor for adiponectin levels.

    C-reactive protein (CRP) is another inflammatory molecule that is affected by oxidative stress andis associated with inflammation and atherosclerosis. It is an acute phase protein that is induced bythe pro-inflammatory cytokine IL-6 and primarily secreted by the liver but also by other cell types.In endothelial cells it induces the expression of adhesion molecules [151] and inhibits endotheliumdependent NO-mediated dilatation [152]. Moreover, in many epidemiological studies it was

    identified as a strong predictor of coronary heart disease and of future cardiovascular events [153,154]. These effects on endothelial cells can induce endothelial dysfunction and atherotrombosis,indicating that it is not simply an inflammatory marker but rather an active participant ininflammatory/atherosclerotic processes [151].

    As with adiponectin it is not conclusive whether CRP levels are elevated in OSAS. Thus far CRP wasreported to increase in a severity dependent manner [155] and to decrease with nCPAP [141].However, additional studies implicated obesity, rather than OSAS, in elevating CRP levels [156160].Thus, as with circulating levels of inflammatory cytokines, it is difficult to tease out the independentcontribution of obesity from OSAS in increasing CRP levels.

    Oxidative stress/inflammation in OSAS and aggregatedcomorbidities

    In recent years it has become increasingly evident that a variety of metabolic risk factors andmorbidities, such as obesity, hypertension, hyperlipidemia and diabetes, are associated withoxidative stress and inflammation. In fact, these metabolic disorders are the very same risk factorsthat cluster with OSAS. Thus, in OSAS there could be a confluence of different potentiallyindependent sources of ROS, which could greatly amplify its effects and contribute to thedevelopment of cardiovascular morbidity as described previously [22]. The central role of ROS

    and the associations with comorbidities are depicted in figure 4. Therefore, in conditions likeobesity and morbidities,e.g.hypertension, hyperlipidemia, or insulin resistance, as in OSAS; thereis a common denominator in the form of oxidative stress/inflammation that promotes endothelialdysfunction and atherosclerosis. In some of these morbidities it is not always clear which factor isthe initiator. Such morbidities could develop independently of OSAS due to the geneticbackground, hormonal, dietary or lifestyle-related variables that greatly affect metabolicdysregulations and obesity. However, it is also likely that such morbidities could develop inmany instances due to OSAS and their consequences as described in figure 4. These mayaccelerate the development of hypertension, hyperlipidemia, insulin resistance, and diabetes [22].It is also very likely that obesity could act as an initiating factor. However, regardless of which

    precedes which, i.e. whether it is OSAS or metabolic dysregulation, once OSAS develops andclusters with components of metabolic dyregulation the nightly initiated oxidative stress turns outto be a key factor in inducing the chain of events that promotes atherosclerosis and eventuallycardiovascular morbidities. It is noteworthy that since most OSAS are obese, apart from the IHwhich is characteristic of OSAS, the subjects are also exposed to continuous sustained hypoxia.This, since sustained hypoxia, is a major feature of the adipose tissue in the obese [161]. Therefore,

    372

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    14/21

    sustained hypoxia and intermit-tent hypoxia may act synergisti-cally to exacerbate the oxidativestress but may also activate differ-ent ROS-dependent signallingpathways [25]. Additional inter-actions of obesity and oxidative

    stress have been described pre-viously [162].

    Inter-individualdifferences in hypoxicstimulation: a targetfor intervention?

    Although OSAS has been re-peatedly demonstrated to be anindependent risk factor for car-diovascular morbidity, not allpatients develop cardiovascularcomplications. These individualdifferences merit a special atten-tion. One highly likely possibilityresides in the existence of large,individual differences in the re-

    sponse of an individual to a givenhypoxia stimulus. These differ-ences may be of such magnitudeand importance as to whether anindividual will withstand a suddenmyocardial infarction, developatherosclerosis or remain free of cardiovascular complications. Although the existing informationregarding individual differences in OSAS is very limited it warrants special attention. Several linesof evidence point to this possibility. First, as previously discussed, some discrepancies wereobserved at the level of the HIF transcription factor and its gene products in models of IH and

    OSAS [90, 9496, 99102]. Such discrepancies could stem from inter-individual differences in theresponse to a given hypoxia stimulus. A study in support of this concept was conducted onpatients with coronary artery disease [163]. In that study, the levels of VEGF produced by monocytesof these patients were measured before and after an identical hypoxic stimulus. There was a largeindividual variability in the VEGF-fold induction among patients. Yet, this increase in VEGF levelswas highly correlated with angiographically documented collateral formation. Such findings indicatethat there are individuals who are high responders to hypoxia and therefore, more prone toproduce VEGF and other vascular growth factors and to develop collateral circulation around theheart, as opposed to low responders with low collateralisation. Additional evidence supporting thisassumption is based on the response of peripheral blood lymphocytes of healthy individuals exposed

    to hypoxia [164]. In this study, the expression of four HIF regulated genes was followed after exposureto various oxygen concentrations. A significant variation was found within the cellular response tohypoxia between normal humans. It was suggested that the common nature of the variability across allfour HIF-regulated genes resides within the HIF system itself. Evidence that variations in HIF-1agenotype influence the development of coronary artery collaterals in patients with significantcoronary artery diseases, indeed suggests a genetic basis for these individual difference [165].

    Dyslipidaemia

    Independent

    HTNDMDyslipidaemia

    Obesity

    Oxidative stress

    SA

    DM

    Inflammation

    Cardiovascularmorbidity

    HTN

    OSAS

    Figure 4. A schematic illustration suggestive of the central roleplayed by oxidative stress and inflammation in obstructive sleepapnoea and the development of associated conditions and

    comorbidities. Associated conditions and comorbidities can be

    induced by oxidative stress or develop independently. Once theseconditions and comorbidities develop, regardless of the initiating

    factors, they elicit a series of intricate interactions with various

    transduction pathways, promoting oxidative stress and inflammation.

    The enhanced oxidative stress exacerbates inflammation, which inturn further exacerbates oxidative stress, generating a vicious cycle

    of oxidative stress and inflammation, eventually leading to cardio-

    vascular morbidity. OSAS: obstructive sleep apnoea syndrome; SA:sympathetic activation; HTN: hypertension; DM: type 2 diabetes

    mellitus. Reproduced from [25] with the permission from the

    publisher.

    373

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    15/21

    Collectively, these findings could explain the inconsistencies in measuring various HIF-1a geneproducts in OSAS.

    The first evidence that OSAS could be associated with an active adaptive mechanism wasprovided by a recent study conducted on patients undergoing elective coronary angiography whowere also screened for OSAS [166]. Interestingly, coronary collateralisation was more developedin the patients with OSAS who had at least one blocked artery, than in their matchedcounterparts without OSAS [166, 167]. Collaterals may protect the myocardium from infarction

    and ischaemia by providing alternative routes of blood supply to regions of the heart supplied byoccluded coronary arteries. Yet, the ability of an individual to develop coronary collateralcirculation is highly variable between patients. It is, therefore, likely that genetic variations in theHIF system of patients with OSAS will result in genetically determined variations in thesensitivity to hypoxia. Thus, patients with a higher sensitivity to the lack of oxygen are morelikely to develop coronary collaterals in response to the nocturnal IH, as opposed to those withlower sensitivity. Several studies conducted on animal models treated with IH are in line with thehuman collaterals findings reported by STEINER et al. [166]. For instance, isolated rat heartstreated with IH were protected against post-ischaemic injury. Moreover, this cardiac protectionwas critically dependent on the dosage of HIF-1a gene [168]. However, several factors, such as

    the depth, the duration and the intermittency of the hypoxia, are crucial and should be explored[169]. In accordance with this line, treatment of mice with IH also resulted in an increase in leftventricular contractility [170]. Thus, the data of STEINERet al. [166] indicate the presence of anadaptive mechanism in the elderly OSAS patient. These findings may also explain some of theclinical observations regarding the age decline trends in mortality of OSAS patients [7, 171].Undoubtedly, the potentially protective qualities of IH in OSAS should be further investigated asthey may have important clinical implications for treatment. Moreover, such findings, ifcombined with individual gene analysis, and personalised medicine, may provide new treatmentstrategies for cardiovascular protection.

    Conclusion

    In conclusion the key important factors to remember when understanding oxidative stress andinflammation in obstructive sleep apnoea patients are as follows. 1) OSAS is an independent riskfactor for cardiovascular morbidity and mortality. 2) It is characterised by intermittent recurrentpauses in respiration termed IH and therefore it is considered as an in vivomodel of hypoxia/reoxygenation. 3) In response to the IH a vicious cycle of oxidative stress/inflammation ensues inOSAS, resulting in endothelial dysfunction and atherosclerosis. 4) Conditions and morbidities thatcluster with OSAS are characterised with an oxidative stress/inflammation component andendothelial dysfunction. These may exacerbate atherosclerotic sequelae. 5) Oxidative stress inOSAS, which results from increased production of ROS and/or decreased antioxidant capacity,alters the redox balance and is likely to activate redox sensitive transcription factors. 6) Thesetranscription factors activate inflammatory and adaptive pathways. 7) Activation of theinflammatory pathways is characterised by increased expression of adhesion molecules and pro-inflammatory cytokines, which promote adhesion and cytotoxicity of various leukocytesubpopulations to endothelial cells. 8) The chain of events characteristic of IH, which promotesatherosclerotic sequelae, can result in cardiovascular morbidity and mortality. However it could beexacerbated by the presence of obesity and metabolic dysregulation. 9) Treatment of OSAS withnCPAP aborts some of these atherosclerotic sequelae. 10) IH may confer beneficial effects in someindividuals in the form of increased collateral circulation.

    Support Statement

    This study was supported in part by a grant from the Binational US-Israel foundation grant#1006695.

    374

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    16/21

    Statement of Interest

    None declared.

    References1. Somers VK, White DP, Amin R,et al. Sleep apnea and cardiovascular disease: an American Heart Association

    American College Of Cardiology Foundation Scientific Statement from the American Heart Association Council for

    High Blood Pressure Research Professional Education Committee, Council on Clinical Cardiology, Stroke Council,and Council On Cardiovascular Nursing. In collaboration with the National Heart, Lung, and Blood Institute

    National Center on Sleep Disorders Research (National Institutes of Health). Circulation2008; 118: 10801111.

    2. Lavie L. Obstructive sleep apnoea syndromean oxidative stress disorder.Sleep Med Rev2003; 7: 3551.

    3. Young T, Palta M, Dempsey J,et al. The occurrence of sleep-disordered breathing among middle-aged adults.

    N Engl J Med1993; 328: 12301235.

    4. Young T, Shahar E, Nieto FJ,et al. Predictors of sleep-disordered breathing in community-dwelling adults: the

    Sleep Heart Health Study. Arch Intern Med2002; 162: 893900.

    5. Punjabi NM. The epidemiology of adult obstructive sleep apnea. Proc Am Thorac Soc2008; 5: 136143.

    6. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective.Am

    J Respir Crit Care Med2002; 165: 12171239.

    7. Lavie P, Lavie L, Herer P. All-cause mortality in males with sleep apnoea syndrome: declining mortality rates with

    age.Eur Respir J2005; 25: 514520.8. Drager LF, Bortolotto LA, Maki-Nunes C,et al.The incremental role of obstructive sleep apnoea on markers of

    atherosclerosis in patients with metabolic syndrome. Atherosclerosis2010; 208: 490495.

    9. Nieto FJ, Peppard PE, Young TB. Sleep disordered breathing and metabolic syndrome.WMJ2009; 108: 263265.

    10. Levy P, Bonsignore MR, Eckel J. Sleep, sleep-disordered breathing and metabolic consequences.Eur Respir J2009;

    34: 243260.

    11. Riha RL, Diefenbach K, Jennum P,et al.Genetic aspects of hypertension and metabolic disease in the obstructive

    sleep apnoea-hypopnoea syndrome. Sleep Med Rev2008; 12: 4963.

    12. Dempsey JA, Veasey SC, Morgan BJ,et al. Pathophysiology of sleep apnea. Physiol Rev2010; 90: 47112.

    13. Somers VK, Mark AL, Abboud FM. Sympathetic activation by hypoxia and hypercapnia-implications for sleep

    apnea.Clin Exp Hypertens A 1988; 10: Suppl. 1, 413422.

    14. Hedner J, Darpo B, Ejnell H,et al. Reduction in sympathetic activity after long-term CPAP treatment in sleep

    apnoea: cardiovascular implications. Eur Respir J1995; 8: 222229.15. Valko M, Leibfritz D, Moncol J,et al. Free radicals and antioxidants in normal physiological functions and

    human disease. Int J Biochem Cell Biol2007; 39: 4484.

    16. Grisham MB, Granger DN, Lefer DJ. Modulation of leukocyte-endothelial interactions by reactive metabolites of

    oxygen and nitrogen: relevance to ischemic heart disease. Free Radic Biol Med1998; 25: 404433.

    17. Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly.Am J

    Physiol1996; 271: C1424C1437.

    18. Suzuki YJ, Forman HJ, Sevanian A. Oxidants as stimulators of signal transduction.Free Radic Biol Med1997; 22:

    269285.

    19. DAutreaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS

    homeostasis. Nat Rev Mol Cell Biol2007; 8: 813824.

    20. Droge W. Free radicals in the physiological control of cell function.Physiol Rev2002; 82: 4795.

    21. Lavie L. Intermittent hypoxia: the culprit of oxidative stress, vascular inflammation and dyslipidemia inobstructive sleep apnea. Expert Rev Resp Med2008; 2: 7584.

    22. Lavie L. Oxidative stress-a unifying paradigm in obstructive sleep apnea and comorbidities.Prog Cardiovasc Dis

    2009; 51: 303312.

    23. Itoh K, Mimura J, Yamamoto M. Discovery of the negative regulator of Nrf2, Keap1: a historical overview.

    Antioxid Redox Signal2010; 13: 16651678.

    24. Libby P. Inflammation in atherosclerosis.Nature2002; 420: 868874.

    25. Lavie L, Lavie P. Molecular mechanisms of cardiovascular disease in OSAHS: the oxidative stress link.Eur Respir J

    2009; 33: 14671484.

    26. Peng YJ, Yuan G, Ramakrishnan D, et al. Heterozygous HIF-1a deficiency impairs carotid body-mediated

    systemic responses and reactive oxygen species generation in mice exposed to intermittent hypoxia. J Physiol

    2006; 577: 705716.

    27. Granger DN, Rutili G, McCord JM. Superoxide radicals in feline intestinal ischemia.Gastroenterology1981; 81:2229.

    28. McCord JM. The evolution of free radicals and oxidative stress.Am J Med2000; 108: 652659.

    29. Lefer DJ, Granger DN. Oxidative stress and cardiac disease.Am J Med2000; 109: 315323.

    30. Peng YJ, Overholt JL, Kline D,et al. Induction of sensory long-term facilitation in the carotid body by

    intermittent hypoxia: implications for recurrent apneas. Proc Natl Acad Sci USA 2003; 100: 1007310078.

    375

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    17/21

    31. Yuan G, Adhikary G, McCormick AA,et al.Role of oxidative stress in intermittent hypoxia-induced immediate

    early gene activation in rat PC12 cells. J Physiol2004; 557: 773783.

    32. Kumar GK, Rai V, Sharma SD,et al.Chronic intermittent hypoxia induces hypoxia-evoked catecholamine efflux

    in adult rat adrenal medulla viaoxidative stress. J Physiol2006; 575: 229239.

    33. Shan X, Chi L, Ke Y,et al. Manganese superoxide dismutase protects mouse cortical neurons from chronic

    intermittent hypoxia-mediated oxidative damage. Neurobiol Dis2007; 28: 206215.

    34. McGown AD, Makker H, Elwell C,et al. Measurement of changes in cytochrome oxidase redox state during

    obstructive sleep apnea using near-infrared spectroscopy. Sleep2003; 26: 710716.

    35. Zhan G, Serrano F, Fenik P,et al. NADPH oxidase mediates hypersomnolence and brain oxidative injury in a

    murine model of sleep apnea. Am J Respir Crit Care Med2005; 172: 921929.36. Peng YJ, Nanduri J, Yuan G,et al. NADPH oxidase is required for the sensory plasticity of the carotid body by

    chronic intermittent hypoxia. J Neurosci2009; 29: 49034910.

    37. Zhan G, Fenik P, Pratico D,et al. Inducible nitric oxide synthase in long-term intermittent hypoxia:

    hypersomnolence and brain injury. Am J Respir Crit Care Med2005; 171: 14141420.

    38. Dyugovskaya L, Lavie P, Lavie L. Increased adhesion molecules expression and production of reactive oxygen

    species in leukocytes of sleep apnea patients. Am J Respir Crit Care Med2002; 165: 934939.

    39. Dyugovskaya L, Polyakov A, Lavie P,et al.Delayed neutrophil apoptosis in patients with sleep apnea. Am J Respir

    Crit Care Med2008; 177: 544554.

    40. Schulz R, Mahmoudi S, Hattar K,et al.Enhanced release of superoxide from polymorphonuclear neutrophils in

    obstructive sleep apnea. Impact of continuous positive airway pressure therapy. Am J Respir Crit Care Med2000;

    162: 566570.

    41. El Solh AA, Saliba R, Bosinski T,et al.Allopurinol improves endothelial function in sleep apnoea: a randomisedcontrolled study. Eur Respir J2006; 27: 9971002.

    42. Saito H, Nishimura M, Shibuya E,et al. Tissue hypoxia in sleep apnea syndrome assessed by uric acid and

    adenosine. Chest2002; 122: 16861694.

    43. Burckhardt IC, Gozal D, Dayyat E,et al. Green tea catechin polyphenols attenuate behavioral and oxidative

    responses to intermittent hypoxia. Am J Respir Crit Care Med2008; 177: 11351141.

    44. Kheirandish L, Gozal D, Pequignot JM, et al. Intermittent hypoxia during development induces long-term

    alterations in spatial working memory, monoamines, and dendritic branching in rat frontal cortex. Pediatr Res

    2005; 58: 594599.

    45. Jun J, Savransky V, Nanayakkara A,et al.Intermittent hypoxia has organ-specific effects on oxidative stress. Am J

    Physiol Regul Integr Comp Physiol2008; 295: R1274R1281.

    46. Peng YJ, Rennison J, Prabhakar NR. Intermittent hypoxia augments carotid body and ventilatory response to

    hypoxia in neonatal rat pups. J Appl Physiol2004; 97: 20202025.47. Park AM, Suzuki YJ. Effects of intermittent hypoxia on oxidative stress-induced myocardial damage in mice.

    J Appl Physiol2007; 102: 18061814.

    48. Lavie L, Vishnevsky A, Lavie P. Evidence for lipid peroxidation in obstructive sleep apnea.Sleep2004; 27:

    123128.

    49. Barcelo A, Miralles C, Barbe F,et al.Abnormal lipid peroxidation in patients with sleep apnoea.Eur Respir J2000;

    16: 644647.

    50. Christou K, Moulas AN, Pastaka C,et al. Antioxidant capacity in obstructive sleep apnea patients. Sleep Med

    2003; 4: 225228.

    51. Tan KC, Chow WS, Lam JC,et al. HDL dysfunction in obstructive sleep apnea. Atherosclerosis2006; 184:

    377382.

    52. Kizawa T, Nakamura Y, Takahashi S,et al. Pathogenic role of angiotensin II and oxidised LDL in obstructive

    sleep apnoea. Eur Respir J2009; 34: 13901398.53. Yamauchi M, Nakano H, Maekawa J, et al. Oxidative stress in obstructive sleep apnea. Chest2005; 127:

    16741679.

    54. Carpagnano GE, Kharitonov SA, Resta O,et al.8-Isoprostane, a marker of oxidative stress, is increased in exhaled

    breath condensate of patients with obstructive sleep apnea after night and is reduced by continuous positive

    airway pressure therapy. Chest2003; 124: 13861392.

    55. Itzhaki S, Dorchin H, Clark G,et al.The effects of 1-year treatment with a herbst mandibular advancement splint

    on obstructive sleep apnea, oxidative stress, and endothelial function. Chest2007; 131: 740749.

    56. Monneret D, Pepin JL, Godin-Ribuot D,et al. Association of urinary 15-F2t-isoprostane level with oxygen

    desaturation and carotid intima-media thickness in nonobese sleep apnea patients.Free Radic Biol Med2010; 48:

    619625.

    57. Pialoux V, Hanly PJ, Foster GE,et al. Effects of exposure to intermittent hypoxia on oxidative stress and acute

    hypoxic ventilatory response in humans. Am J Respir Crit Care Med2009; 180: 10021009.

    58. Hoffmann MS, Singh P, Wolk R,et al.Microarray studies of genomic oxidative stress and cell cycle responses in

    obstructive sleep apnea. Antioxid Redox Signal2007; 9: 661669.

    59. Barcelo A, Barbe F, de la Pena M,et al.Antioxidant status in patients with sleep apnoea and impact of continuous

    positive airway pressure treatment. Eur Respir J2006; 27: 756760.

    60. Faure P, Tamisier R, Baguet JP,et al.Impairment of serum albumin antioxidant properties in obstructive sleep

    apnoea syndrome.Eur Respir J2008; 31: 10461053.

    376

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    18/21

    61. Lavie L, Hefetz A, Luboshitzky R,et al.Plasma levels of nitric oxide and L-arginine in sleep apnea patients: effects

    of nCPAP treatment. J Mol Neurosci2003; 21: 5763.

    62. Schulz R, Schmidt D, Blum A,et al. Decreased plasma levels of nitric oxide derivatives in obstructive sleep

    apnoea: response to CPAP therapy. Thorax2000; 55: 10461051.

    63. Ip MS, Lam B, Chan LY,et al.Circulating nitric oxide is suppressed in obstructive sleep apnea and is reversed by

    nasal continuous positive airway pressure. Am J Respir Crit Care Med2000; 162: 21662171.

    64. Lavie L. Sleep-disordered breathing and cerebrovascular disease: a mechanistic approach.Neurol Clin2005; 23:

    10591075.

    65. Jelic S, Padeletti M, Kawut SM, et al. Inflammation, oxidative stress, and repair capacity of the vascular

    endothelium in obstructive sleep apnea. Circulation2008; 117: 22702278.66. Jelic S, Lederer DJ, Adams T,et al. Vascular inflammation in obesity and sleep apnea. Circulation2010; 121:

    10141021.

    67. Grebe M, Eisele HJ, Weissmann N,et al. Antioxidant vitamin C improves endothelial function in obstructive

    sleep apnea. Am J Respir Crit Care Med2006; 173: 897901.

    68. de Lima AM, Franco CM, de Castro CM,et al.Effects of nasal continuous positive airway pressure treatment on

    oxidative stress and adiponectin levels in obese patients with obstructive sleep apnea. Respiration2010; 79: 370376.

    69. Carlson JT, Rangemark C, Hedner JA. Attenuated endothelium-dependent vascular relaxation in patients with

    sleep apnoea. J Hypertens1996; 14: 577584.

    70. Kato M, Roberts-Thomson P, Phillips BG,et al.Impairment of endothelium-dependent vasodilation of resistance

    vessels in patients with obstructive sleep apnea. Circulation2000; 102: 26072610.

    71. Itzhaki S, Lavie L, Pillar G, et al. Endothelial dysfunction in obstructive sleep apnea measured by peripheral

    arterial tone response in the finger to reactive hyperemia. Sleep2005; 28: 594600.72. Nagahama H, Soejima M, Uenomachi H, et al. Pulse wave velocity as an indicator of atherosclerosis in

    obstructive sleep apnea syndrome patients. Intern Med2004; 43: 184188.

    73. Minoguchi K, Yokoe T, Tazaki T,et al. Increased carotid intima-media thickness and serum inflammatory

    markers in obstructive sleep apnea. Am J Respir Crit Care Med2005; 172: 625630.

    74. Drager LF, Bortolotto LA, Lorenzi MC,et al.Early signs of atherosclerosis in obstructive sleep apnea. Am J Respir

    Crit Care Med2005; 172: 613618.

    75. Friedlander AH, Yueh R, Littner MR. The prevalence of calcified carotid artery atheromas in patients with

    obstructive sleep apnea syndrome. J Oral Maxillofac Surg1998; 56: 950954.

    76. Jelic S, Bartels MN, Mateika JH,et al.Arterial stiffness increases during obstructive sleep apneas.Sleep2002; 25:

    850855.

    77. Drager LF, Bortolotto LA, Figueiredo AC,et al. Effects of continuous positive airway pressure on early signs of

    atherosclerosis in obstructive sleep apnea. Am J Respir Crit Care Med2007; 176: 706712.78. Ip MS, Tse HF, Lam B, et al. Endothelial function in obstructive sleep apnea and response to treatment. Am J

    Respir Crit Care Med2004; 169: 348353.

    79. Wan F, Lenardo MJ. The nuclear signaling of NF-kappaB: current knowledge, new insights, and future

    perspectives.Cell Res2010; 20: 2433.

    80. Karin M, Takahashi T, Kapahi P,et al. Oxidative stress and gene expression: the AP-1 and NF-kB connections.

    Biofactors2001; 15: 8789.

    81. Pierce GL, Lesniewski LA, Lawson BR, et al. Nuclear factor-kB activation contributes to vascular endothelial

    dysfunctionviaoxidative stress in overweight/obese middle-aged and older humans. Circulation2009; 119:

    12841292.

    82. Htoo AK, Greenberg H, Tongia S,et al. Activation of nuclear factor kB in obstructive sleep apnea: a pathway

    leading to systemic inflammation. Sleep Breath2006; 10: 4350.

    83. Greenberg H, Ye X, Wilson D,et al. Chronic intermittent hypoxia activates nuclear factor-k

    B in cardiovasculartissuesin vivo. Biochem Biophys Res Commun2006; 343: 591596.

    84. Yamauchi M, Tamaki S, Tomoda K,et al.Evidence for activation of nuclear factor kB in obstructive sleep apnea.

    Sleep Breath2006; 10: 189193.

    85. Dyugovskaya L, Polyakov A, Ginsberg D,et al. Molecular pathways of spontaneous and TNF-a-mediated

    neutrophil apoptosis under intermittent hypoxia. Am J Respir Cell Mol Biol2010; [Epub ahead of print DOI:

    10.1165/RCMB.2010-0025OC].

    86. Ryan S, Taylor CT, McNicholas WT. Predictors of elevated nuclear factor-kappaB-dependent genes in obstructive

    sleep apnea syndrome.Am J Respir Crit Care Med2006; 174: 824830.

    87. Lavie L, Polotsky V. Cardiovascular aspects in obstructive sleep apnea syndrome - molecular issues, hypoxia and

    cytokine profiles. Respiration2009; 78: 361370.

    88. Dyugovskaya L, Lavie P, Lavie L. Lymphocyte activation as a possible measure of atherosclerotic risk in patients

    with sleep apnea. Ann N Y Acad Sci2005; 1051: 340350.

    89. Lavie L, Dyugovskaya L, Lavie P. Sleep-apnea-related intermittent hypoxia and atherogenesis: adhesion molecules

    and monocytes/endothelial cells interactions. Atherosclerosis2005; 183: 183184.

    90. Ryan S, Taylor CT, McNicholas WT. Selective activation of inflammatory pathways by intermittent hypoxia in

    obstructive sleep apnea syndrome. Circulation2005; 112: 26602667.

    91. Rey S, Semenza GL. Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular

    remodelling.Cardiovasc Res2010; 86: 236242.

    377

    L.

    LAVIE

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    19/21

    92. Semenza GL, Prabhakar NR. HIF-1-dependent respiratory, cardiovascular, and redox responses to chronic

    intermittent hypoxia. Antioxid Redox Signal2007; 9: 13911396.

    93. Prabhakar NR, Kumar GK, Nanduri J. Intermittent hypoxia-mediated plasticity of acute O2 sensing requires

    altered red-ox regulation by HIF-1 and HIF-2. Ann N Y Acad Sci2009; 1177: 162168.

    94. Yuan G, Nanduri J, Bhasker CR,et al.Ca2+/calmodulin kinase-dependent activation of hypoxia inducible factor

    1 transcriptional activity in cells subjected to intermittent hypoxia. J Biol Chem 2005; 280: 43214328.

    95. Toffoli S, Feron O, Raes M,et al. Intermittent hypoxia changes HIF-1alpha phosphorylation pattern in

    endothelial cells: unravelling of a new PKA-dependent regulation of HIF-1alpha. Biochim Biophys Acta2007;

    1773: 15581571.

    96. Polotsky VY, Savransky V, Bevans-Fonti S,et al. Intermittent and sustained hypoxia induce a similar geneexpression profile in the human aortic endothelial cells. Physiol Genomics2010; 41: 306314.

    97. Belaidi E, Joyeux-Faure M, Ribuot C,et al.Major role for hypoxia inducible factor-1 and the endothelin system

    in promoting myocardial infarction and hypertension in an animal model of obstructive sleep apnea. J Am Coll

    Cardiol2009; 53: 13091317.

    98. Semenza GL. Oxygen-regulated transcription factors and their role in pulmonary disease.Respir Res2000; 1: 159162.

    99. Li J, Bosch-Marce M, Nanayakkara A, et al. Altered metabolic responses to intermittent hypoxia in mice with

    partial deficiency of hypoxia-inducible factor-1a. Physiol Genomics2006; 25: 450457.

    100. Winnicki M, Shamsuzzaman A, Lanfranchi P,et al.Erythropoietin and obstructive sleep apnea. Am J Hypertens

    2004; 17: 783786.

    101. Lavie L, Kraiczi H, Hefetz A,et al.Plasma vascular endothelial growth factor in sleep apnea syndrome: effects of

    nasal continuous positive air pressure treatment. Am J Respir Crit Care Med2002; 165: 16241628.

    102. Valipour A, Litschauer B, Mittermayer F,et al.Circulating plasma levels of vascular endothelial growth factor inpatients with sleep disordered breathing. Respir Med2004; 98: 11801186.

    103. Wakabayashi N, Slocum SL, Skoko JJ, et al.When NRF2 talks, whos listening? Antioxid Redox Signal2010; 13:

    16491663.

    104. Malec V, Gottschald OR, Li S,et al.HIF-1a signaling is augmented during intermittent hypoxia by induction of

    the Nrf2 pathway in NOX1-expressing adenocarcinoma A549 cells. Free Radic Biol Med2010; 48: 16261635.

    105. Belaiba RS, Bonello S, Zahringer C,et al. Hypoxia up-regulates hypoxia-inducible factor-1a transcription by

    involving phosphatidylinositol 3-kinase and nuclear factor kappaB in pulmonary artery smooth muscle cells. Mol

    Biol Cell2007; 18: 46914697.

    106. Walmsley SR, Print C, Farahi N, et al. Hypoxia-induced neutrophil survival is mediated by HIF-1a-dependent

    NF-kappaB activity. J Exp Med2005; 201: 105115.

    107. Zhou J, Damdimopoulos AE, Spyrou G, et al. Thioredoxin 1 and thioredoxin 2 have opposed regulatory

    functions on hypoxia-inducible factor-1a. J Biol Chem 2007; 282: 74827490.108. Taylor CT, Cummins EP. The role of NF-kB in hypoxia-induced gene expression.Ann N Y Acad Sci2009; 1177:

    178184.

    109. Panes J, Granger DN. Leukocyte-endothelial cell interactions: molecular mechanisms and implications in

    gastrointestinal disease. Gastroenterology1998; 114: 10661090.

    110. Dyugovskaya L, Lavie P, Hirsh M,et al.Activated CD8+T-lymphocytes in obstructive sleep apnoea.Eur Respir J

    2005; 25: 820828.

    111. Dyugovskaya L, Lavie P, Lavie L. Phenotypic and functional characterization of blood gammadelta T cells in sleep

    apnea.Am J Respir Crit Care Med2003; 168: 242249.

    112. McDonald PP. Transcriptional regulation in neutrophils: teaching old cells new tricks.Adv Immunol2004; 82: 148.

    113. Libby P. Inflammatory mechanisms: the molecular basis of inflammation and disease.Nutr Rev2007; 65:

    S140S146.

    114. Minoguchi K, Tazaki T, Yokoe T,et al.Elevated production of tumor necrosis factor-a

    by monocytes in patientswith obstructive sleep apnea syndrome. Chest2004; 126: 14731479.

    115. Vanderlaan PA, Reardon CA. Thematic review series: the immune system and atherogenesis. The unusual

    suspects: an overview of the minor leukocyte populations in atherosclerosis. J Lipid Res2005; 46: 829838.

    116. Song L, Leung C, Schindler C. Lymphocytes are important in early atherosclerosis.J Clin Invest2001; 108:

    251259.

    117. Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-interactions: linking hemostasis and

    inflammation. Blood Rev2007; 21: 99111.

    118. Bokinsky G, Miller M, Ault K,et al. Spontaneous platelet activation and aggregation during obstructive sleep

    apnea and its response to therapy with nasal continuous positive airway pressure. A preliminary investigation.

    Chest1995; 108: 625630.

    119. Geiser T, Buck F, Meyer BJ,et al. In vivoplatelet activation is increased during sleep in patients with obstructive

    sleep apnea syndrome. Respiration2002; 69: 229234.

    120. Eisensehr I, Ehrenberg BL, Noachtar S,et al.Platelet activation, epinephrine, and blood pressure in obstructive

    sleep apnea syndrome. Neurology1998; 51: 188195.

    121. Hui DS, Ko FW, Fok JP, et al. The effects of nasal continuous positive airway pressure on platelet activation in

    obstructive sleep apnea syndrome. Chest2004; 125: 17681775.

    122. Nobili L, Schiavi G, Bozano E, et al. Morning increase of whole blood viscosity in obstructive sleep apnea

    syndrome. Clin Hemorheol Microcirc2000; 22: 2127.

    378

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    20/21

  • 8/14/2019 Oxidative stress and inflammation in OSA.pdf

    21/21

    155. Shamsuzzaman AS, Winnicki M, Lanfranchi P,et al.Elevated C-reactive protein in patients with obstructive sleep

    apnea.Circulation2002; 105: 24622464.

    156. Ryan S, Nolan GM, Hannigan E,et al. Cardiovascular risk markers in obstructive sleep apnoea syndrome and

    correlation with obesity. Thorax2007; 62: 509514.

    157. Guilleminault C, Kirisoglu C, Ohayon MM. C-reactive protein and sleep-disordered breathing. Sleep2004; 27:

    15071511.

    158. Taheri S, Austin D, Lin L,et al.Correlates of serum C-reactive protein (CRP)no association with sleep duration

    or sleep disordered breathing. Sleep2007; 30: 991996.

    159. Sharma SK, Mishra HK, Sharma H, et al. Obesity, and not obstructive sleep apnea, is responsible for increased

    serum hs-CRP levels in patients with sleep-disordered breathing in Delhi. Sleep Med2008; 9: 149156.160. Lavie L, Vishnevsky A, Lavie P. Oxidative stress and systemic inflammation in patients with sleep apnea: role of

    obesity.Sleep Biol Rhythm 2007; 5: 100110.

    161. Trayhurn P, Wang B, Wood IS. Hypoxia in adipose tissue: a basis for the dysregulation of tissue function in

    obesity? Br J Nutr2008; 100: 227235.

    162. Vincent HK, Taylor AG. Biomarkers and potential mechanisms of obesity-induced oxidant stress in humans.Int

    J Obes (Lond)2006; 30: 400418.

    163. Schultz A, Lavie L, Hochberg I, et al. Interindividual heterogeneity in the hypoxic regulation of VEGF:

    significance for the development of the coronary artery collateral circulation. Circulation1999; 100: 547552.

    164. Brooks JT, Elvidge GP, Glenny L, et al. Variations within oxygen-regulated gene expression in humans. J Appl

    Physiol2009; 106: 212220.

    165. Resar JR, Roguin A, Voner J, et al. Hypoxia-inducible factor 1a polymorphism and coronary collaterals in

    patients with ischemic heart disease. Chest2005; 128: 787791.166. Steiner S, Schueller PO, Schulze V, et al. Occurrence of coronary collateral vessels in patients with sleep apnea

    and total coronary occlusion. Chest2010; 137: 516520.

    167. Lavie L, Lavie P. Coronary collateral circulation in sleep apnea: a cardioprotective mechanism? Chest2010; 137:

    511512.

    168. Cai Z, Manalo DJ, Wei G, et al. Hearts from rodents exposed to intermittent hypoxia or erythropoietin are

    protected against ischemia-reperfusion injury. Circulation2003; 108: 7985.

    169. Beguin PC, Belaidi E, Godin-Ribuot D,et al.Intermittent hypoxia-induced delayed cardioprotection is mediated

    by PKC and triggered by p38 MAP kinase and Erk1/2. J Mol Cell Cardiol2007; 42: 343351.

    170. Naghshin J, McGaffin KR, Witham WG,et al.Chronic intermittent hypoxia increases left ventricular contractility

    in C57BL/6J mice. J Appl Physiol2009; 107: 787793.

    171. Lavie P, Lavie L. Unexpected survival advantage in elderly people with moderate sleep apnoea.J Sleep Res2009;

    18: 397403.

    OXIDATIVE

    STRESS

    AND

    INFLAMM

    ATION