You are on page 1of 10

Review

For reprint orders, please contact:


reprints@future-drugs.com

Intermittent hypoxia: the


culprit of oxidative stress,
vascular inflammation and
dyslipidemia in obstructive
sleep apnea
Expert Rev. Resp. Med. 2(1), 75–84 (2008)

Lena Lavie Obstructive sleep apnea, a breathing disorder in sleep characterized by intermittent and
Unit of Anatomy and Cell recurrent pauses in respiration, is also a major risk factor for cardiovascular morbidity and
Biology, The Ruth and Bruce mortality. Accumulated evidence implicates the apnea-related multiple cycles of
Rappaport Faculty of hypoxia/reoxygenation in promoting the formation of reactive oxygen species that oxidize and
Medicine, Technion, damage macromolecules and activate critical redox-sensitive signaling pathways and
PO Box 9649, 31096, transcription factors. This activation facilitates the expression of sets of genes encoding
Haifa, Israel proteins in various pathways, including inflammatory and lipogenic, as well as proteins
Tel.:+972 4829 5234 essential to adaptation to hypoxia. Consequently, inflammatory and immune responses are
Fax: +972 4834 3934/ activated, thus resulting in the activation of endothelial cells/leukocytes/platelets. These
4829 5403 activated cells express adhesion molecules and proinflammatory cytokines that in turn may
lenal@tx.technion.ac.il further exacerbate inflammatory responses and cause endothelial cell injury and dysfunction,
promoting the development of cardiovascular morbidities in sleep apnea. No less important in
activating such inflammatory cascades is the hyperlipidemia that is another characteristic of
obstructive sleep apnea. Evidence supporting the existence of endothelial dysfunction and
early clinical signs of atherosclerosis in these patients provides a firm support to the above
chain of events. If left untreated, this cascade of events may eventually lead to overt
cardiovascular morbidity.

KEYWORDS: atherosclerosis • endothelial cells • hyperlipidemia • inflammation • leukocytes • obstructive sleep apnea
• oxidative stress • platelets

In 1993, based mainly on theoretical consid- to the underlying pathophysiology of cardio-


erations and the knowledge available at the vascular morbidity in sleep apnea [4,5]. Only in
time, the foundation of the ‘free radi- the early 2000s was the first direct evidence
cal/inflammatory theory in sleep apnea’ was demonstrating increased oxidative stress in OSA
laid [1]. In this seminal paper, Dean and Wil- published [6,7] and indirect evidence substanti-
cox suggested that the repeated breathing ated it [8,9]. By 2003, in a comprehensive review
arrests accompanied by drastic changes in by Lavie [10] based on the same theoretical con-
PO 2, which are the hallmark of obstructive siderations and on newly acquired data perti-
sleep apnea (OSA) syndrome, are analogous to nent to hypoxia/reoxygenation biology of leuko-
hypoxia and reoxygenation and may lead to cytes/endothelial cell interactions [11] and on
free radical bursts, as was demonstrated in sporadic data on sleep apnea, the links
conditions of ischemia and reperfusion [2]. between atherosclerosis and OSA were further
Based on this assumption of free radical produc- elaborated [10].
tion in sleep apnea, combined with Ross’s Since then, more than 80 publications on oxi-
hypothesis of ‘the response to injury’ describing dative stress and more than 100 on inflamma-
the development of atherosclerosis [3], this prop- tion and sleep apnea have been published and
osition provided a very attractive explanation provided unequivocal support to this hypothesis.

www.future-drugs.com 10.1586/17476348.2.1.75 © 2008 Future Drugs Ltd ISSN 1747-6348 75


Review Lavie

Several mechanisms were proposed to delineate the associa- risk factor for cardiovascular morbidity [31], while Lavie sum-
tion between OSA and cardiovascular morbidity. These marized the evidence on the risk of mortality in sleep apnea
included increases in sympathetic nerve activity, swings in patients [32].
intrathoracic pressure, altered blood coagulability and, as out-
lined previously, the participation of atherogenic sequelae and
their underlying mechanisms that are uniquely triggered by OSA & dyslipidemia
apnea-related intermittent hypoxia (IH) [4] and resultant oxi- Hyperlipidemia is also a prevalent finding among sleep apnea
dative stress [10,12,13]. These include inflammation and possi- patients. In a multiethnic cohort of 6440 men and women of the
bly hyperlipidemia, which is a well-established risk factor for Sleep Health Heart Study, Newman et al. demonstrated an
cardiovascular morbidity. increase in total serum cholesterol and triglycerides levels and a
The present paper summarizes the current knowledge on decrease in high-density lipoproteins (HDLs) independent of age
atherosclerosis in OSA. It describes the underlying fundamental and BMI [33]. Similar data were reported on a matched control
mechanisms of oxidative stress leading to cellular inflammatory study of sleep apnea patients [34]. Moreover, effective ameliora-
and immune activation as well as the associated dyslipidemia. tion of the apneas with nasal continous positive airway pressure
(nCPAP) treatment lowered serum total cholesterol levels [35–37].
In accord with this line, HDL of patients with OSA was shown
OSA & cardiovascular morbidity to be dysfunctional by lacking the ability to inactivate oxidized
Sleep apnea syndrome is characterized by intermittent and lipids [38]. This HDL dysfunction can be at least partially attrib-
recurrent pauses in respiration that result in decreases in uted to the lower activity of paraoxonase-1 detected in patients
blood oxygen saturation and sleep fragmentation. OSA is a with OSA, particularly in those who also have cardiovascular
prevalent syndrome [5], one in four men and one in ten comorbidities [39]. Paraoxonase-1 is an antioxidant enzyme that
women have at least five disordered breathing events in each is located exclusively on HDL and protects both low-density
hour of sleep in the form of apneas or hypopneas [14]. Apneas lipoprotein (LDL) and HDL from oxidative modifications.
are complete cessations of breathing for at least a 10-s dura- Thus, lower activities of paraoxonase-1 render patients with OSA
tion, while hypopneas are partial cessations for the same dura- susceptible to HDL dysfunction.
tion (defined as apnea–hypopnea index [AHI], denoting the
number of breathing arrests per hour of sleep). In 4% of adult
men and 2% of adult women, sleep apnea syndrome is accom- Oxidative stress: its sources & consequences in OSA
panied by characteristic symptoms, such as excessive daytime Oxidative stress
sleepiness, chronic fatigue or neurocognitive decline, which Oxidative stress is characterized by an imbalance between oxi-
comprise the sleep apnea syndrome [14]. Specifically, the syn- dant-producing systems and antioxidant defense mechanisms
drome is associated with the male gender, middle age, central (redox balance), which results in excessive formation of reactive
obesity, smoking, sedentary lifestyle and a postmenopausal oxygen species (ROS). It represents a common threat and a haz-
status in women [15]. ard to all aerobic organisms, as it may severely influence the
At the time of diagnosis, at least 50% of sleep apnea progress of various pathological conditions. The predominant
-
patients are hypertensive and 10–15% have cardiovascular ROS molecule is the superoxide anion (O2• ) radical. It is gener-
disease, including myocardial infarction and stroke [16,17]. On ated by univalent reduction of molecular oxygen mainly during
the other hand, breathing disorders in sleep are prevalent respiration, but also by several other enzymic systems:
- -
among patients with cardiovascular diseases [18,19]. These O2 + e →O2•
-
cross-sectional data on the association between cardiovascular Once formed, the O2• can be dismutated spontaneously or
morbidity and breathing disorders in sleep are supported by by the superoxide dismutase (SOD) enzyme. This reaction
large population-based studies demonstrating that this associ- yields molecular oxygen and hydrogen peroxide (H2O2):
- -
ation is independent of all possible confounding cardiovascu- O2• + O2• + 2H+ →O2 + H2O2
•-
lar risk factors [20–22] and by prospective studies [23]. There is When O2 reacts with its product of dismutation (H2O2), a
particularly strong evidence supporting a causal relationship hydroxyl radical (OH• ), which is a highly oxidant molecule, can
between sleep apnea and hypertension, which is based on be formed. The formation of OH• is facilitated by the presence
large epidemiological studies [16,20–22], prospective studies [23], of reduced transition metals such as Fe3+ and Cu+2 [40]:
- -
the effect of treatment that ameliorated the apnea on blood O2• + H2O2 →O2 + OH• + OH
-
pressure [24] and on an animal model of sleep apnea [25]. Sev- Collectively, these molecules (O2• , H2O2 and OH• ) are
eral studies also demonstrated that sleep apnea syndrome is termed ROS.
an independent risk factor for cardiovascular mortality [26–28] Additional toxic radicals, termed reactive nitrogen species
and that effective treatment of the syndrome significantly (RNS), also contribute to oxidative/nitrosative stress, such as the
-
decreased mortality [29,30]. McNicholas et al. provided a peroxynitrite (OONO ). This molecule is formed by the reaction
•-
recent up-to-date review on sleep apnea as an independent of O2 with the primary vasodilator nitric oxide (NO):

76 Expert Rev. Resp. Med. 2(1), (2008)


Intermittent hypoxia in obstructive sleep apnea Review

- -
O2• + NO →OONO responses that ultimately lead to atherosclerosis [54]. In partic-
-
The presence of OONO drastically affects endothelial ular, ROS were implicated in initiation of inflammatory path-
function as it diminishes NO availability [41]. ways via activation of nuclear transcription factors, such as
nuclear factor (NF)κB and activator protein (AP)-1, which
Pathways of increased ROS formation in OSA orchestrate the production of adhesion molecules and inflam-
Generally, one of the main sources of ROS formation is from matory cytokines. Another group of transcription factors that
mitochondria through electron leakage during respiration, which could possibly be affected by redox imbalance and oxidative
serves for signaling purposes. It is estimated that, under normal stress are the sterol regulator element-binding proteins
physiological conditions, approximately 5% of the inspired oxy- (SREBPs) [55]. These SREBPs that activate genes regulating
gen is converted to ROS. During hypoxia/reoxygenation condi- lipid metabolism [56,57] were shown to be upregulated in
tions, mitochondria become dysfunctional [42]. However, forma- experimental models of IH [58,59].
tion of ROS can result from activation of various ROS- Induction of the master regulator hypoxia-inducible factor
producing enzymes, such as xanthine oxidase and NADPH oxi- (HIF)-1α, which is essential for oxygen homeostasis and
dase, from activated leukocytes and endothelial cells [11,43]. adaptive response to hypoxia, was documented primarily in
Additional possible sources include enzymes such as cyclooxyge- several experimental models of IH in tissue culture and
nase, lipooxyganase, NO synthase and heme oxygenases [12]. In rodents exposed to chronic IH [60]. Moreover, delineating the
OSA, mitochondrial dysfunction was demonstrated through transduction signals that activate HIF-1α under IH condi-
measurement of changes in the cytochrome oxidase redox state tions were found to differ from sustained hypoxia [61].
during obstructive apnea [44]. Activation of xanthine oxidase was Although HIF-1α activation was not directly demonstrated in
implicated indirectly through increases in the levels of metabolic patients with OSA, upregulation of some of its gene products,
by-products that characterize this pathway [2]. Thus, increases in including erythropoietin, VEGF and heat-shock proteins,
by-products such as uric acid and adenosine were reported in supports this notion [10,62,63].
OSA [10]. However, the data implicating increased activity of Additional redox-sensitive transcription factors with far-
NADPH oxidase are the most convincing. The NADPH oxidase reaching implications to cardiovascular morbidity that merit
is the primary enzyme that is utilized by inflammatory phagocytic intensive investigation in OSA include members of the GATA
cells to generate ROS in order to kill invading microorganisms. family. Emerging evidence implicates GATA-4 and -6 in regula-
However, ROS are also produced by phagocytes at low levels tion of cardiac development and growth as well as in heart fail-
under basal conditions [45]. In patients with OSA, both mono- ure. Altered balance of these transcription factors may cause car-
cytes and granulocytes were shown to be activated and to release diac hypertrophy or promote cardioprotection [64]. In recent
two- to threefold higher amounts of ROS compared with control studies headed by Suzuki and colleagues utilizing a mouse
subjects [6,7]. In mice exposed to IH conditions, NADPH oxidase model of IH, GATA-4 was shown to exert preconditioning-like
was implicated in brain injury [46]. The result of such increases in cardioprotective effects by protecting cardiomyocytes from
ROS formation in OSA, as well as in animal models treated by apoptosis. However, while acute IH was cardioprotective, expo-
IH, is evident by the presence of oxidized macromolecules, such as sure of mice to prolonged IH resulted in increased susceptibility
lipids, proteins DNA and carbohydrates [8,9,39,46–50]. In parallel, of the heart via increased oxidative stress. Further exposure to
sleep apnea patients also demonstrate decreased antioxidant prolonged IH facilitated reversal of the enhancement of myo-
capacities [51,52,39], which further disrupt the tightly regulated cardial damage [65,66]. Thus, elucidating the complex effects of
cellular oxidation–reduction (redox) state. IH on these transcription factors in the human heart could be
Collectively, the wealth of data accumulated thus far clearly helpful in identifying patients with OSA at risk of developing
attest to the presence of oxidative stress in OSA by increased cardiovascular morbidity or, alternatively, cardioprotection.
production of ROS as well as by decreased antioxidant capacity.

Consequences of oxidative stress NFκB & inflammatory pathway activation in


A large body of evidence demonstrates that ROS molecules sleep apnea
play a dual role. On the one hand, due to their high chemical Activation of the inflammatory pathways via NFκB upregu-
reactivity, as mentioned previously, ROS can damage various lation was recently demonstrated in neutrophils and mono-
biomolecules and cellular components, such as lipids, proteins, cytes of patients with OSA [67–69], as well as in a tissue cul-
carbohydrates and DNA, by oxidation and, by that, can alter ture model utilizing HeLa cells that were exposed to IH
their functions and induce various pathologies. On the other in vitro [70]. Also, increased AP-1 activity and tyrosine
hand, under normal physiological conditions, ROS function as hydroxylase mRNA, an AP-1-regulated downstream gene,
signaling molecules [53]. Thus, the altered ROS balance may were shown in PC12 cells exposed to IH [71]. Accordingly,
activate some signaling pathways and inhibit others, leading to upregulation of adhesion molecules and inflammatory
altered gene expression. Consequently, the plethora of signaling cytokines, the gene products of NFκB and AP-1, in patients
pathways that are activated can initiate inflammatory with OSA further support NFκB and AP-1 activation [10,12].

www.future-drugs.com 77
Review Lavie

Moreover, IH in vitro was shown to activate NFκB in an IκB of venous (HUVEC) and arterial origin (HCAEC) was noted.
kinase (IKK)-dependent manner at least in part via activation Moreover, by utilizing antibodies to selectins (anti-CD62) and
of p38 MAPK [72]. integrins (anti-CD54), the adhesion to endothelial cells was
Thus, the ROS molecules produced in response to IH by the attenuated to values comparable with controls [7].
ROS-producing enzymes and dysfunctional mitochondria, pre-
sumably from endothelial cells and leukocytes, initiate a cascade Cytotoxic T lymphocytes
of inflammatory pathways resulting in overexpression of adhesion The cytotoxic T-lymphocyte subpopulation of patients with OSA
molecules and proinflammatory cytokines. These adhesion mole- that were investigated also expressed an activated and cytotoxic
cules facilitate the recruitment and accumulation of leukocytes phenotype. Increased adhesion to endothelial cells and increased
and platelets on the endothelial cells lining the vasculature and cytotoxicity towards endothelial cells were shown by the CD8+,
promote neutrophils/monocyte/lymphocyte/platelets/endothelial CD4+ and γ δ-cytotoxic T lymphocytes compared with controls
+
cell interactions. Such cellular interactions between blood cells [73–75]. Moreover, the killing abilities of CD8 T lymphocytes
and endothelial cells may result in injury to the endothelium. were found to also be AHI-severity dependent. Yet, each sub-
population employed different killing mechanisms to damage
Adhesion molecules on leukocytes & interactions with endothelial cells [73–75]. While CD8+ T lymphocytes primarily
endothelial cells expressed higher amounts of the CD56 natural killer receptors and
Adhesion molecules are upregulated in endothelial cells and perforin, CD4+ killing relied on the presence of CD4+/CD28null T
blood leukocytes in response to various pathological conditions cells and the killing by γ δ+ T lymphocytes was primarily mediated
and insults, such as hypoxia/reoxygenation. Their expression is by the presence of the proinflammatory cytokine TNF-α, which
a highly regulated, sequential process and is exhibited in both was found to be elevated in patients with OSA.
endothelial cells and leukocytes and promotes the interactions
between these cell types. These sequential interactions involve Platelets
three independent steps: rolling, firm adhesion and trans- Similarly to circulating leukocytes, platelets have also been
migration [54]. In each step, a defined set of adhesion molecules shown to acquire an activated and a prothrombotic phenotype
participate, having specific receptors and counter-receptors on in response to hypoxia/reoxygenation. Platelets from patients
leukocytes and endothelial cells [54]. The interactions between with OSA expressed increased activation and aggregability
endothelial cells and various leukocyte subpopulations of in vitro. The percentage of platelets expressing P-selectin
patients with OSA, including monocytes, granulocytes and (CD62P) was higher [78,79], mainly in the severe group of
numerous cytotoxic T cells expressing CD8, CD4 and γ δ, were patients [80] and was effectively lowered by treatment with
rigorously investigated in our laboratory [7,73–77]. nCPAP [81]. In addition, increases in hematocrit, blood viscosity
and fibrinogen in patients with OSA could further affect hyper-
Neutrophils coagulability [82,83]. Furthermore, in an ongoing study in our
The expression of the CD15 carbohydrate complex on selectins (of lab, we found that platelets of patients with OSA form higher
the family of adhesion molecules that mediate rolling) of OSA amounts of platelets/monocytes aggregates compared with their
neutrophils was increased in a severity-dependent manner. Treat- age-, sex- and BMI-matched controls (VISHNEVSKY A, LAVIE P, LAVIE L,
ment with nCPAP downregulated its expression. On the other UNPUBLISHED OBSERVATIONS ). Owing to the fact that platelets were

hand, the expression of CD11c, a β-subunit of the integrins (and a shown to play a key role in ischemic cardiovascular diseases,
counter receptor for intercellular adhesion molecule (ICAM)-1 on their altered activation state and hyperaggregability may also
endothelial cells responsible for firm adhesion), was unaffected. contribute to increased cardiovascular morbidity in OSA.
Furthermore, neutrophil apoptosis – a fundamental injury-limit- Collectively, the higher expression of adhesion molecules on
ing mechanism that prevents its destructive potential – was leukocytes, their higher avidity and the ability to strongly attach
delayed in OSA; thus, together with increased expression of adhe- to endothelial cells in culture conditions, the stronger cyto-
sion molecules, delayed neutrophil apoptosis may promote toxicity of T cells against endothelial cells, the delayed apoptosis
endothelial injury [77]. expressed by neutrophils, the higher ROS generated by mono-
cytes and neutrophils, and the higher aggregability of platelets
Monocytes are all markers of activation of the various blood cells investi-
The expression of two adhesion molecules was increased in mono- gated in patients with OSA and can serve as markers of the pos-
cytes of patients with OSA compared with controls. Notably, sible ongoing processes that may damage the endothelium and
CD15 and CD11c were elevated, while treatment with nCPAP initiate atherogenesis.
attenuated their expression [7]. Moreover, increased CD15 expres-
sion on monocytes was dependent on the severity of the syn- Activated endothelial cells in OSA
drome [76] and treatment of monocytes from healthy individuals The occurrence of soluble variants of adhesion molecules origi-
with hypoxia in vitro upregulated their expression. Thus, increased nating in endothelial cells were found in plasma or sera of
adhesion of monocytes from patients with OSA to endothelial cells patients with OSA. These variant adhesion molecules are shed

78 Expert Rev. Resp. Med. 2(1), (2008)


Intermittent hypoxia in obstructive sleep apnea Review

when endothelial cells become activated. Several well-estab- lipid peroxidation and atherosclerosis were both dependent on
lished markers were identified, including P- and E-selectin, the severity of the chronic IH and that the SREBP-1 pathway
ICAM-1 and vascular cell adhesion molecule (VCAM)-1. All mediated the hyperlipdemia observed in this model [86,89].
are considered as markers of active atherosclerotic diseases and Given that OSA is associated with hyperlipidemia independ-
as predictors of future cardiovascular disease [12]. ent of obesity, as manifested by various studies [33–37] and
based on the above cited animal studies, it is reasonable to
Proinflammatory cytokines assume that the hyperlipidemia observed in OSA is mediated
Proinflammatory cytokines, similar to adhesion molecules, via upregulation of the SREBP-1 pathway. No less important
are affected by the redox state of the cells in which they are is the fact that oxidative stress was implicated in the upregula-
synthesized and actively participate and modulate inflam- tion of SREBP, while this upregulation was sensitive to anti-
matory responses. Cytokines are multipurpose molecules and oxidants [55]. Thus, IH and oxidative stress in humans, as in
regulate both the innate and adaptive immune system. They experimental models, may upregulate SREBP-1, leading to the
regulate macrophage activation via expression of scavenger hyperlipidemia associated with OSA.
receptors and secretion of metalloproteinases, modulate the
proliferation of smooth muscle cells, the production of nitric
oxide and apoptosis, and stimulate the activation of endothe- Endothelial dysfunction & early signs
lial cells: all of which are steps in the progression of athero- of atherosclerosis
sclerosis. The main cytokines investigated in OSA include In the normal state, the endothelium is responsible for regula-
TNF-α, IL-6 and IL-8, and the anti-inflammatory cytokine tion of the vascular tone and interactions between the vessel wall
IL-10, which were all shown to be affected by OSA. Evi- and circulating substances and blood cells. Thus, it maintains an
dently, once the inflammatory response is initiated, these anticoagulant and anti-inflammatory phenotype. Upon disrup-
cytokines can in turn activate NFκB and, by that, further tion of this balance, the endothelium is activated and can
exaggerate inflammation. In patients with OSA, increases in acquire a prothrombotic and proinflammatory phenotype that
proinflammatory cytokine levels were primarily found in the leads to endothelial dysfunction – the earliest sign of atheroscle-
circulation [84]. However, elevated levels in monocytes (GOLAN- rosis [54,90]. Basically, endothelial dysfunction represents a state
SHANY O, LAVIE P, LAVIE L, UNPUBLISHED OBSERVATIONS) and in various cyto- in which vasodilatation of the blood vessels is compromised due
toxic T lymphocytes were also observed [74,75]. Critically, to decreased NO bioavailability. NO is the most potent vasodi-
patients with sleep apnea, besides having increased expression lator but it also mediates many of the protective functions of the
of proinflammatory cytokines, also display an altered proin- endothelium. It limits leukocyte recruitment and leukocyte
flammatory/anti-inflammatory cytokine balance. For expression of adhesion molecules. It prevents proliferation of
instance, CD8+ T lymphocytes expressed higher levels of vascular smooth muscle cells and platelet aggregation and adhe-
TNF-α than controls. However, the levels of the anti-inflam- sion. Thus, it protects from atherosclerosis. Exposure to oxida-
matory cytokine IL-10 were also increased, presumably as a tive stress, inflammatory mediators or hypercholesterolemia, all
protective mechanism to counteract TNF-α actions; yet, this of which promote endothelial cell activation and endothelial
increase in IL-10 levels was not proportional to the increases dysfunction, impair NO bioavailability [54,90,91]. In patients with
obtained in the production of TNF-α [75]. OSA, several studies have demonstrated diminished NO bioa-
vailability, which was restored after nCPAP treatment [92–94].
These data further implicate atherosclerotic sequlae in the devel-
Sterol regulator element-binding proteins opment of cardiovascular morbidity in OSA. In addition, several
Another possible set of transcription factors that could be a measures that represent early signs of atherosclerosis were also
plausible candidate for the growing list of redox-regulated tran- shown to be elevated in OSA, including increased intima–media
scription factors in OSA is the family of SREBPs. The SREBPs thickness that was also severity dependent [95–97], arterial plaque
are endoplasmic reticulum membrane-bound transcription fac- formation [98], calcified artery atheromas [99] and higher pulse
tors that activate more then 30 genes encoding the enzymes wave velocity [97,100]. Thus, cardiovascular morbidity in sleep
that regulate the synthesis and uptake of cholesterol, fatty acids, apnea is foreseeable. Moreover, this is further demonstrated by a
triglycerides and phospholipids [57]. The activity of SREBPs is recent study showing that such early signs of atherosclerosis were
regulated by sterol feedback inhibition. Normally, SREBP is significantly improved by 4-month treatment with nCPAP [95].
activated in sterol-depleted cells and is inactivated with high However, these recent findings should also be reproduced and
cholesterol levels. However, under IH conditions, there could investigated in patients of moderate severity.
be an over-ride of this mechanism due to lack of oxygen, which This course of events, starting with sleep apnea IH and oxidative
leads to upregulation of SREBPs [85–87]. stress through activation of cellular inflammatory pathways and
Recently, in a series of elegant studies, Polotsky’s group pathways that cause hyperlipidemia, may injure the endothe-
established the development of atherosclerosis in a rodent lium and consequently impose cardiovascular morbidity or,
model exposed to chronic IH [88]. This group also found that alternatively, may confer cardioprotection, as illustrated in FIGURE 1.

www.future-drugs.com 79
Review Lavie

OSA/IH

ROS

Redox-sensitive transcription factor activation

??*
NFκB, AP-1 SREBP HIF-1a

??
Inflammation Hyperlipidemia Adaptation
GATA-4
• Adhesion molecules • Cholesterol and fatty acid synthesis • VEGF
• Cytokines • Cellular uptake of lipoproteins • EPO
• Leukocytes Protection • HSPs
• Platelets
• Endothelial cells
Endothelial dysfunction

Atherosclerosis

Cardiovascular morbidity Cardioprotection?


Cardiac hypertrophy?

Figure 1. Accelerated atherosclerosis and development of cardiovascular morbidity in OSA. IH initiates activation of ROS-
producing systems, resulting in an altered redox balance and activating various redox-sensitive transcription factors. Activation of
the inflammatory pathway via NFκB leads to activation of various leukocyte subpopulations (neutrophils, monocytes and
lymphocytes), platelets and endothelial cells, facilitating increased injury to endothelial cells via adhesion molecules,
proinflammatory cytokines, further increases in ROS molecules and diminished NO bioavailability, culminating in endothelial
dysfunction, the primary step in the development of atherosclerosis. Activation of SREBPs increases lipogenesis and elevates
cholesterol levels in the circulation, which also leads to endothelial dysfunction. The question arises as to whether this mechanism
then acts through inflammatory pathways, further exacerbating them. Adaptive mechanisms are also activated via HIF-1 and
upregulation of its protein products and may protect some individuals more than others. Altered expression of GATA-4, a cardiac
muscle cell survival transcription factor, may directly promote either cardioprotection or development of cardiac hypertrophy.
AP: Activator protein; EPO: Erythropoietin; HIF: Hypoxia-inducible factor; HSP: Heat-shock protein; OSA: Obstructive sleep apnea;
ROS: Reactive oxygen species; SREBP: Sterol regulator element-binding protein; VEGF: Vascular endothelial growth factor.
*
So far supported by induction via oxidative stress induced by Hepatitis C virus [55].

diseases by the time of diagnosis. It is evident that mecha-


Expert commentary nisms such as increased oxidative stress, activation of redox-
In recent years, a large body of evidence implicates oxidative sensitive transcription factors and the inflammatory/immune
stress, inflammation and, more recently, hyperlipidemia as cells, accompanied by expression of various adhesion mole-
fundamental mechanisms in the pathophysiology of cardio- cules and inflammatory cytokines that culminate on endothe-
vascular morbidity in OSA. These confirmed the identifica- lial dysfunction, share many similarities with atherogenic
tion of OSA as an independent risk factor for cardiovascular pathways of ischemia/reperfusion injury and most likely with
morbidity. As outlined in this review, the sequence of events hypercholesterolemia as well.
starting with the nightly occurrence of IH and ending with Some of these early clinical signs are dependent on apnea
endothelial dysfunction provides an explanation of why OSA severity and could be reversed by treatment with nCPAP.
patients, free of any overt cardiovascular diseases, have dis- These early clinical signs of atherosclerosis most probably
played early signs of atherosclerosis as increased carotid start to accrue during the early stages of the disease, regard-
intima–media thickness, arterial plaque formation, calcified less of patient’s symptoms. Patients, however, are generally
artery atheromas, atherosclerotic lesions and higher pulse referred for diagnosis only when the characteristic symptoms
wave velocity, and why many of them display cardiovascular of snoring or excessive daytime sleepiness start to affect them

80 Expert Rev. Resp. Med. 2(1), (2008)


Intermittent hypoxia in obstructive sleep apnea Review

or are noticed by people around them, generally around the The pathophysiology of IH on the major metabolic organ –
age of 50 years. Diagnosis and treatment of OSA at this age, the liver – is only just being unveiled. Currently, the unknown
which is delayed by 10–20 years from the time damage to the exceeds by far the knowledge accumulated; this is also true for
cardiovascular system was incited, may be too late. Further- the heart. It is very likely that, in the next few years, the effects
more, as mortality risk in sleep apnea is maximal at younger of IH will be expanded to additional internal organs, such as
ages [101], it is also too late for many of the patients who are at the brain, kidneys, spleen, endocrine organs and muscles.
maximal risk of dying. Thus, one of the most important clin- Moreover, the interactions of OSA and the resultant oxidative
ical lessons learnt from understanding the underlying patho- stress with hypertension, diabetes, hyperlipidemia and obesity
physiology of OSA is that, in order to prevent cardiovascular will be delineated at the cellular and molecular levels.
morbidity, treatment of breathing disorders in sleep should Importantly, as not all patients with sleep apnea develop
start at the earliest possible age. cardiovascular morbidity, it is obvious that OSA not only
elicits inflammatory and hyperlipidemic pathways but also
adaptive pathways that result in ischemic preconditioning of
Five-year view the heart and possibly the brain and liver. The question as to
Sleep medicine has seen unprecedented growth in the last who is at risk of developing cardiovascular morbidity, and if
two decades, mostly owing to the growing awareness of OSA there is a genetic basis for this adaptation, will be addressed in
and its profound impact on patients’ quality of life and the next few years.
cardiovascular health. In the next few years, based on the Finally, in the last 25 years, treatment of OSA almost
mechanistic insights accumulated in OSA and from the exclusively relies on the nCPAP, which was shown to be effec-
ongoing studies on in vivo and in vitro models of IH, tive in reversing endothelial dysfunction and preventing car-
research will further elaborate and refine the understanding diovascular sequela. In the next few years, research will focus
of the cellular and molecular mechanisms mediating the on examining the efficacy of other treatment modalities, such
effects of IH on the cardiovascular system. Specifically, I as more effective and more friendly user alternatives to
believe that the importance of nitrosative mechanisms that nCPAP, dental devices or massive weight reduction. In view
induce nitrosative stress under IH conditions and additional of the pivotal role played by oxidative stress and hyper-
redox-sensitive transcription factors activated in OSA lipidemia in this process, the possible effects of antioxidants
patients, by IH, will be identified. Specific transduction and statins will also be explored.
pathways of the redox-activated transcription factors will
most likely be elucidated with their similarities/dis-
similarities to mechanisms of sustained hypoxia and to Financial & competing interests disclosure
ischemia/reperfusion already established. Likewise, the The author has no relevant affiliations or financial involvement with any
sequence of interactions of various leukocytes with the organization or entity with a financial interest in or financial conflict
endothelium exposed to IH in animal models in vivo will be with the subject matter or materials discussed in the manuscript. This
clarified using various knockout and transgenic animal mod- includes employment, consultancies, honoraria, stock ownership or options,
els exposed to IH and utilizing methods such as intravital expert testimony, grants or patents received or pending, or royalties.
microscopy. No writing assistance was utilized in the production of this manuscript.

Key issues
• Obstructive sleep apnea is an independent risk factor for cardiovascular morbidity and mortality.
• The intermittent recurrent pauses in respiration in sleep apnea, in a similar manner to hypoxia/reoxygenation, induce oxidative stress
and diminish nitric oxide availability.
• Increased production of reactive oxygen species (ROS) and/or decreased antioxidant capacity that result in oxidative stress and altered
redox balance activate redox-sensitive transcription factors.
• These transcription factors activate inflammatory, hyperlipidemic and adaptive pathways but may also directly damage the heart or
induce cardioprotection.
• Activation of the inflammatory pathways, characterized by increased expression of adhesion molecules and proinflammatory
cytokines, leads to an increase in adhesion and cytotoxicity by various leukocyte subpopulations towards endothelial cells.
• Activation of lipogenic pathways by intermittent hypoxia results in hypercholesterolemia. This may exaggerate ongoing inflammatory
pathways already activated by the intermittent hypoxia.
• The above chain of events results in atherosclerotic sequelae, such as endothelial dysfunction and early signs of atherosclerosis,
which, if untreated, will eventually manifest in overt cardiovascular diseases.
• Treatment of sleep apnea should be initiated as early as possible in order to abort the atherosclerotic chain of events.

www.future-drugs.com 81
Review Lavie

13 Suzuki YJ, Jain V, Park AM, Day RM. 24 Pepperell JC, Ramdassingh-Dow S,
References Oxidative stress and oxidant signaling in Crosthwaite N et al. Ambulatory blood
Papers of special note have been highlighted as: obstructive sleep apnea and associated pressure after therapeutic and subtherapeutic
• of interest cardiovascular diseases. Free Radic. Biol. Med. nasal continuous positive airway pressure for
•• of considerable interest 40, 1683–1692 (2006). obstructive sleep apnoea: a randomised
14 Young T, Palta M, Dempsey J, Skatrud J, parallel trial. Lancet 359, 204–210 (2002).
1 Dean RT, Wilcox I. Possible atherogenic
effects of hypoxia during obstructive sleep Weber S, Badr S. The occurrence of sleep- 25 Brooks D, Horner RL, Kozar LF,
apnea. Sleep 16(8 Suppl.), S15–S22 (1993). disordered breathing among middle-aged Render-Teixeira CL, Phillipson EA.
adults. N. Engl. J. Med. 328, 1230–1235 Obstructive sleep apnea as a cause of systemic
2 McCord JM. Oxygen-derived free radicals in
(1993). hypertension. Evidence from a canine model.
postischemic tissue injury. N. Engl. J. Med.
•• Seminal paper about the prevalence of J. Clin. Invest. 99, 106–109 (1997).
312(3), 159–163 (1985).
sleep apnea. 26 He J, Kryger MH, Zorick FJ, Conway W,
3 Ross R. The pathogenesis of atherosclerosis –
15 Young T, Shahar E, Nieto FJ et al. Predictors Roth T. Mortality and apnea index in
an update. N. Engl. J. Med. 314(8), 488–500
obstructive sleep apnea. Experience in 385
(1986). of sleep-disordered breathing in community-
male patients. Chest 94, 9–14 (1988).
dwelling adults: the Sleep Heart Health
4 Wolk R, Somers VK. Cardiovascular
Study. Arch. Intern. Med. 162, 893–900 27 Lavie P, Herer P, Peled R et al. Mortality in
consequences of obstructive sleep apnea.
(2002). sleep apnea patients: a multivariate analysis of
Clin. Chest Med. 24, 195–205 (2003).
16 Lavie P, Herer P, Hoffstein V. Obstructive risk factors. Sleep 18, 149–157 (1995).
5 Malhotra A, White DP. Obstructive sleep
sleep apnoea syndrome as a risk factor for 28 Yaggi HK, Concato J, Kernan WN,
apnoea. Lancet 360, 237–245 (2002).
hypertension: population study. BMJ 320, Lichtman JH, Brass LM, Mohsenin V.
•• Excellent review on sleep apnea. 479–482 (2000). Obstructive sleep apnea as a risk factor for
6 Schulz R, Mahmoudi S, Hattar K et al. stroke and death. N. Engl. J. Med. 353,
• Important paper describing the relationship
Enhanced release of superoxide from 2034–2041 (2005).
between sleep apnea and hypertension in a
polymorphonuclear neutrophils in clinical population. 29 Marti S, Sampol G, Munoz X et al. Mortality
obstructive sleep apnea. Impact of continuous in severe sleep apnoea/hypopnea syndrome
positive airway pressure therapy. Am. J. Respir. 17 Smith R, Ronald J, Delaive K, Walld R,
patients: impact of treatment. Eur. Respir. J.
Crit. Care Med. 162, 566–570 (2000). Manfreda J, Kryger MH. What are
20, 1511–1518 (2002).
obstructive sleep apnea patients being treated
7 Dyugovskaya L, Lavie P, Lavie L. Increased for prior to this diagnosis? Chest 121, 30 Marin JM, Carrizo SJ, Vicente E, Agusti AG.
adhesion molecules expression and 164–172 (2002). Long-term cardiovascular outcomes in men
production of reactive oxygen species in with obstructive sleep apnoea–hypopnoea
leukocytes of sleep apnea patients. Am. J. 18 Peker Y, Kraiczi H, Hedner J, Löth S,
with or without treatment with continuous
Respir. Crit. Care Med. 165, 934–939 (2002). Johansson A, Bende M. An independent
positive airway pressure: an observational
association between obstructive sleep apnoea
•• Seminal paper on monocyte adhesion study. Lancet 365, 1046–1053 (2005).
and coronary artery disease. Eur. Respir. J. 14,
molecules and respiratory burst activity in 179–184 (1999). 31 McNicholas WT, Bonsigore MR and
sleep apnea. Management Committee of EU COST
19 Mooe T, Rabben T, Wiklund U et al.
8 Saarelainen S, Lehtimaki T, Jaak-kola O et al. ACTION B26. Sleep apnoea as an
Sleep-disordered breathing in men with
Autoantibodies against oxidised low-density independent risk factor for cardiovascular
coronary artery disease. Chest 109, 659–663
lipoprotein in patients with obstructive sleep disease: current evidence, basic mechanisms
(1996).
apnoea. Clin. Chem. Lab. Med. 37(5), and research priorities. Eur. Respir. J. 29,
20 Nieto FJ, Young TB, Lind BK et al. 156–178 (2007).
517–520 (1999).
Association of sleep-disordered breathing,
9 Barcelo A, Miralles C, Barbe F, Vila M, Pons 32 Lavie P. Mortality in sleep apnoea syndrome:
sleep apnea, and hypertension in a large
S, Agusti AG. Abnormal lipid peroxidation in review of the evidence. Eur. Respir. Rev. 16,
community-based study. Sleep Heart Health
patients with sleep apnoea. Eur. Respir. J. 203–210 (2007).
Study. JAMA 283, 1829–1836 (2000).
16(4), 644–647 (2000). 33 Newman AB, Nieto FJ, Guidry U et al.;
21 Bixler EO, Vgontzas AN, Lin HM et al.
10 Lavie L. Obstructive sleep apnoea syndrome Sleep Heart Health Study Research Group.
Association of hypertension and sleep-
– an oxidative stress disorder. Sleep Med. Rev. Relation of sleep-disordered breathing to
disordered breathing. Arch. Intern. Med. 160,
7, 35–51 (2003). cardiovascular disease risk factors: the Sleep
2289–2295 (2000).
Heart Health Study. Am. J. Epidemiol.
•• Comprehensive and thorough review on 22 Young T, Peppard P, Palta M et al. 154(1), 50–59 (2001).
oxidative stress and inflammation in Population-based study of sleep-disordered
34 McArdle N, Hillman D, Beilin L, Watts G.
obstructive sleep apnea. breathing as a risk factor for hypertension.
Metabolic risk factors for vascular disease in
11 Panes J, Granger DN. Leukocyte–endothelial Arch. Intern. Med. 157, 1746–1752 (1997).
obstructive sleep apnea: a matched controlled
cell interactions: molecular mechanisms and 23 Peppard PE, Young T, Palta M et al. study. Am. J. Respir. Crit. Care Med. 175,
implications in gastrointestinal disease. Prospective study of the association between 190–195 (2007).
Gastroenterology 114(5), 1066–1090 (1998). sleep disordered breathing and hypertension.
35 Robinson GV, Pepperell JC, Segal HC,
12 Lavie L. Sleep-disordered breathing and N. Engl. J. Med. 342, 1738–1784 (2000).
Davies RJ, Stradling JR. Circulating
cerebrovascular disease: a mechanistic • First prospective study to demonstrate an cardiovascular risk factors in obstructive sleep
approach. Neurol. Clin. 23, 1059–1075 independent relationship between sleep apnoea: data from randomised controlled
(2005). apnea and increased blood pressure. trials. Thorax 59, 777–782 (2004).

82 Expert Rev. Resp. Med. 2(1), (2008)


Intermittent hypoxia in obstructive sleep apnea Review

36 Chin K, Shimizu K, Nakamura T et al. neuronal cell apoptosis in a mouse model of 60 Semenza GL, Prabhakar NR.
Changes in intra-abdominal visceral fat and sleep apnea. Neuroscience 126, 313–323 HIF-1-dependent respiratory,
serum leptin levels in patients with (2004). cardiovascular, and redox responses to
obstructive sleep apnea syndrome following 49 Minoguchi K, Yokoe T, Tanaka A et al. chronic intermittent hypoxia. Antioxid.
nasal continuous positive airway pressure Association between lipid peroxidation and Redox Signal. 9, 1391–1396 (2007).
therapy. Circulation 100, 706–712 (1999). inflammation in obstructive sleep apnoea. • Important review on hypoxia-inducible
37 Chin K, Nakamura T, Shimizu K et al. Eur. Respir. J. 28, 378–385 (2006). factor-1 intermittent hypoxia and
Effects of nasal continuous positive airway 50 Carpagnano GE, Kharitonov SA, Resta O, cardiovascular sequelae.
pressure on soluble cell adhesion molecules in Foschino-Barbaro MP, Gramiccioni E, 61 Yuan G, Nanduri J, Bhasker CR,
patients with obstructive sleep apnea Barnes PJ. 8-isoprostane, a marker of Semenza GL, Prabhakar NR.
syndrome. Am. J. Med. 109, 562–567 oxidative stress, is increased in exhaled Ca2+/calmodulin kinase-dependent
(2000). breath condensate of patients with activation of hypoxia inducible factor 1
38 Tan KC, Chow WS, Lam JC et al. HDL obstructive sleep apnea after night and is transcriptional activity in cells subjected
dysfunction in obstructive sleep apnea. reduced by continuous positive airway to intermittent hypoxia. J. Biol. Chem.
Atherosclerosis 184, 377–382 (2006). pressure therapy. Chest 124, 1386–1392 280, 4321–4328 (2005).
39 Lavie L, Vishnevsky A, Lavie P. Evidence for (2003).
62 Winnicki M, Shamsuzzaman A,
lipid peroxidation in obstructive sleep apnea. 51 Christou K, Moulas AN, Pastaka C, Lanfranchi P et al. Erythropoietin and
Sleep 27, 123–128 (2004). Gourgoulianis KI. Antioxidant capacity in obstructive sleep apnea. Am. J. Hypertens.
40 Halliwell B, Gutteridge JMC. Role of free obstructive sleep apnea patients. Sleep Med. 17, 783–786 (2004).
radicals and catalytic metal ions in human 27, 997–1002 (2003).
63 Lavie L, Kraiczi H, Hefetz A et al. Plasma
disease: an overview. Meth. Enzymol. 186, 52 Barcelo A, Barbe F, de la Pena M et al. vascular endothelial growth factor in sleep
1–85 (1990). Antioxidant status in patients with sleep apnea syndrome: effects of nasal
41 Beckman JS, Koppenol WH. Nitric oxide, apnoea and impact of continuous positive continuous positive air pressure
superoxide, and peroxynitrite: the good, the airway pressure treatment. Eur. Respir. J. 27, treatment. Am. J. Respir. Crit. Care Med.
bad, and the ugly. Am. J. Physiol. 271, 756–760 (2006). 165, 1624–1628 (2002).
C1421–C1437 (1996). 53 Suzuki YJ, Forman HJ, Sevanian A. 64 Pikkarainen S, Tokola H, Kerkela R,
42 Duranteau J, Chandel NS, Kulisz A, Shao Z, Oxidants as stimulators of signal Ruskoaho H. GATA transcription factors
Schumacker PT. Intracellular signaling by transduction. Free Radic. Biol. Med. 22, in the developing and adult heart.
reactive oxygen species during hypoxia in 269–285 (1997). Cardiovasc. Res. 63, 196–207 (2004).
cardiomyocytes. J. Biol. Chem. 273, 54 Libby P. Inflammation in atherosclerosis. 65 Park AM, Nagase H, Vinod Kumar S,
11619–11624 (1998). Nature 420, 868–874 (2002). Suzuki YJ. Acute intermittent hypoxia
43 Grisham MB, Granger DN, Lefer DJ. • Comprehensive review on inflammation activates myocardial cell survival
Modulation of leukocyte–endothelial in atherosclerosis. signaling. Am. J. Physiol. Heart Circ.
interactions by reactive metabolites of oxygen Physiol. 292, H751–H757 (2007).
55 Waris G, Felmlee DJ, Negro F, Siddiqui A.
and nitrogen: relevance to ischemic heart Hepatitis C virus induces proteolytic 66 Park AM, Nagase H, Kumar SV, Suzuki
disease. Free Radic. Biol. Med. 25, 404–433 YJ. Effects of intermittent hypoxia on the
cleavage of sterol regulatory element binding
(1998). heart. Antioxid. Redox Signal. 9, 723–729
proteins and stimulates their
• Excellent review on cellular interations and phosphorylation via oxidative stress. J. Virol. (2007).
reactive oxygen species. 81, 8122–8130 (2007). 67 Htoo AK, Greenberg H, Tongia S et al.
44 McGown AD, Makker H, Elwell C, 56 Brown MS, Goldstein JL. A proteolytic Activation of nuclear factor κB in
Al Rawi PG, Valipour A, Spiro SG. pathway that controls the cholesterol obstructive sleep apnea: a pathway leading
Measurement of changes in cytochrome content of membranes, cells, and blood. to systemic inflammation. Sleep Breath.
oxidase redox state during obstructive sleep Proc. Natl Acad. Sci. USA 96(20), 10, 43–50 (2006).
apnea using near-infrared spectroscopy. Sleep 11041–11048 (1999). 68 Greenberg H, Ye X, Wilson D, Htoo AK,
26, 710–716 (2003). 57 Horton JD, Goldstein JL, Brown MS. Hendersen T, Liu SF. Chronic
45 Babior BM. Phagocytes and oxidative stress. SREBPs: activators of the complete program intermittent hypoxia activates nuclear
Am. J. Med. 109, 33–44 (2000). of cholesterol and fatty acid synthesis in the factor-κB in cardiovascular tissues in vivo.
liver. J. Clin. Invest. 109, 1125–1131 (2002). Biochem. Biophys. Res. Commun. 343,
46 Zhan G, Serrano F, Fenik P et al. NADPH
591–596 (2006).
oxidase mediates hypersomnolence and brain 58 Li J, Grigoryev DN, Ye SQ et al. Chronic
oxidative injury in a murine model of sleep intermittent hypoxia upregulates genes of 69 Yamauchi M, Tamaki S, Tomoda K et al.
apnea. Am. J. Respir. Crit. Care Med. 172, lipid biosynthesis in obese mice. J. Appl. Evidence for activation of nuclear factor
921–929 (2005). Physiol. 99, 1643–1648 (2005). κB in obstructive sleep apnea. Sleep
Breath. 10, 189–193 (2006).
47 Yamauchi M, Nakano H, Maekawa J et al. 59 Li J, Thorne LN, Punjabi NM et al.
Oxidative stress in obstructive sleep apnea. Intermittent hypoxia induces hyperlipidemia 70 Ryan S, Taylor CT, McNicholas WT.
Chest 127, 1674–1679 (2005). in lean mice. Circ. Res. 97, 698–706 (2005). Selective activation of inflammatory
pathways by intermittent hypoxia in
48 Xu W, Chi L, Row BW et al. Increased • Describes the effects of intermittent
obstructive sleep apnea syndrome.
oxidative stress is associated with chronic hypoxia on the development
Circulation 112, 2660–2667 (2005).
intermittent hypoxia-mediated brain cortical of hyperlipidemia.

www.future-drugs.com 83
Review Lavie

71 Yuan G, Adhikary G, McCormick AA, 82 Hoffstein V, Herridge M, Mateika S, 94 Lavie L, Hefetz A, Luboshitzky R, Lavie P.
Holcroft JJ, Kumar GK, Prabhakar NR. Redline S, Strohl KP. Hematocrit levels in Plasma levels of nitric oxide and L-arginine in
Role of oxidative stress in intermittent sleep apnea. Chest 106, 787–791 (1994). sleep apnea patients: effects of nCPAP
hypoxia-induced immediate early gene 83 Nobili L, Schiavi G, Bozano E, De Carli F, treatment. J. Mol. Neurosci. 21, 57–63
activation in rat PC12 cells. J. Physiol. Ferrillo F, Nobili F. Morning increase of (2003).
557(Pt 3), 773–783 (2004). whole blood viscosity in obstructive sleep 95 Drager LF, Bortolotto LA, Figueiredo AC,
72 Ryan S, McNicholas WT, Taylor CT. apnea syndrome. Clin. Hemorheol. Microcirc. Krieger EM, Lorenzi-Filho G. Effects of
A critical role for p38 map kinase in NF-κB 22, 21–27 (2000). continuous positive airway pressure on Early
signaling during intermittent 84 Vgontzas AN, Papanicolaou DA, Bixler EO Signs of Atherosclerosis in Obstructive Sleep
hypoxia/reoxygenation. Biochem. Biophys. et al. Sleep apnea and daytime sleepiness and Apnea. Am. J. Respir. Crit. Care Med. 176,
Res. Commun. 355, 728–733 (2007). fatigue: relation to visceral obesity, insulin 706–712 (2007).
73 Dyugovskaya L, Lavie P, Hirsh M, Lavie L. resistance, and hypercytokinemia. J. Clin. • Important paper demonstrating reversal of
Activated CD8+ T-lymphocytes in Endocrinol. Metab. 85, 1151–1158 (2000). atherosclerotic signs by nasal continous
obstructive sleep apnoea. Eur. Respir. J. 25, 85 Veasey S. Good soldier falls: the liver in sleep positive airway pressure treatment.
820–828 (2005). apnea. J. Appl. Physiol. 102, 513–514 (2007). 96 Minoguchi K, Yokoe T, Tazaki T et al.
74 Dyugovskaya L, Lavie P, Lavie L. 86 Li J, Nanayakkara A, Jun J, Savransky V, Increased carotid intima–media thickness
Phenotypic and functional characterization Polotsky VY. The effect of deficiency in and serum inflammatory markers in
of blood γ δ T cells in sleep apnea. Am. J. SREBP cleavage-activating protein (SCAP) obstructive sleep apnea. Am. J. Respir. Crit.
Respir. Crit. Care Med. 168, 242–249 on lipid metabolism during intermittent Care Med. 172, 625–630 (2005).
(2003). hypoxia. Physiol. Genomics 31, 273–280 97 Drager LF, Bortolotto LA, Lorenzi MC,
• Describes the contribution of γ / δ T cells (2007). Figueiredo AC, Krieger EM,
to atherosclerotic sequelae in sleep apnea. 87 Hughes AL, Todd BL, Espenshade PJ. Lorenzi-Filho G. Early signs of
75 Dyugovskaya L, Lavie P, Lavie L. SREBP pathway responds to sterols and atherosclerosis in obstructive sleep apnea.
Lymphocyte activation as a possible measure functions as an oxygen sensor in fission yeast. Am. J. Respir. Crit. Care Med. 172, 613–618
of atherosclerotic risk in patients with sleep Cell 120, 831–842 (2005). (2005).
apnea. Ann. NY Acad. Sci. 1051, 340–350 88 Savransky V, Nanayakkara A, Li J et al. 98 Kaynak D, Goksan B, Kaynak H,
(2005). Chronic intermittent hypoxia induces Degirmenci N, Daglioglu S. Is there a link
76 Lavie L, Dyugovskaya L, Lavie P. Sleep atherosclerosis. Am. J. Respir. Crit. Care Med. between the severity of sleep-disordered
apnea related intermittent hypoxia and 175, 1290–1297 (2007). breathing and atherosclerotic disease of the
atherogenesis: adhesion molecules and carotid arteries? Eur. J. Neurol. 10, 487–493
•• Important paper on the effects of
monocytes/endothelial cells interactions. (2003).
intermittent hypoxia on the development
Atherosclerosis 183, 183–184 (2005). of atherosclerosis. 99 Friedlander AH, Yueh R, Littner MR. The
77 Dyugovskaya L, Polyakov A, Lavie P, prevalence of calcified carotid artery
89 Li J, Savransky V, Nanayakkara A, Smith PL,
Lavie L. Delayed neutrophil apoptosis in atheromas in patients with obstructive sleep
O’Donnell CP, Polotsky VY. Hyperlipidemia
sleep apnea patients. Am. J. Respir. Crit. Care apnea syndrome. J. Oral Maxillofac. Surg.
and lipid peroxidation are dependent on the
Med. (2007) (In Press). 56(8), 950–954 (1998).
severity of chronic intermittent hypoxia.
78 Bokinsky G, Miller M, Ault K, Husband P, J. Appl. Physiol. 102, 557–563 (2007). 100 Nagahama H, Soejima M, Uenomachi H
Mitchell J. Spontaneous platelet activation et al. Pulse wave velocity as an indicator of
90 Davignon J, Ganz P. Role of endothelial
and aggregation during obstructive sleep atherosclerosis in obstructive sleep apnea
dysfunction in atherosclerosis. Circulation
apnea and its response to therapy with nasal syndrome patients. Intern. Med. 43, 184–188
109(23 Suppl. 1), III27–III32 (2004).
continuous positive airway pressure. A (2004).
preliminary investigation. Chest 108, 91 Lavie L. Sleep apnea syndrome, endothelial
101 Lavie P, Lavie L, Herer P. All-cause mortality
625–630 (1995). dysfunction, and cardiovascular morbidity.
in males with sleep apnoea syndrome:
Sleep 27, 1053–1055 (2004).
79 Geiser T, Buck F, Meyer BJ, Bassetti C, declining mortality rates with age. Eur. Respir.
Haeberli A, Gugger M. In vivo platelet 92 Ip MS, Lam B, Chan LY et al. Circulating J. 25, 514–520 (2005).
activation is increased during sleep in nitric oxide is suppressed in obstructive sleep
patients with obstructive sleep apnea apnea and is reversed by nasal continuous
syndrome. Respiration 69, 229–234 (2002). positive airway pressure. Am. J. Respir. Crit. Affiliation
Care Med. 162, 2166–2171 (2000).
80 Eisensehr I, Ehrenberg BL, Noachtar S et al. Lena Lavie, PhD
Platelet activation, epinephrine, and blood 93 Schulz R, Schmidt D, Blum A et al. Unit of Anatomy and Cell Biology, The Ruth and
pressure in obstructive sleep apnea Decreased plasma levels of nitric oxide Bruce Rappaport Faculty of Medicine, Technion,
syndrome. Neurology 51, 188–195 (1998). derivatives in obstructive sleep apnoea: PO Box 9649, 31096, Haifa, Israel
response to CPAP therapy. Thorax 55, Tel.: +972 4829 5234
81 Hui DS, Ko FW, Fok JP et al. The effects of 1046–1051 (2000).
nasal continuous positive airway pressure on Fax: +972 4834 3934/4829 5403
platelet activation in obstructive sleep apnea lenal@tx.technion.ac.il
syndrome. Chest 125, 1768–1775 (2004).

84 Expert Rev. Resp. Med. 2(1), (2008)

You might also like