Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 26.
Published in final edited form as: Curr Atheroscler Rep. 2012 Oct;14(5):10.1007/s11883-012-0266-8. doi: 10.1007/s11883-012-0266-8

Evolving Concepts of Oxidative Stress and Reactive Oxygen Species in Cardiovascular Disease

Kai Chen 1, John F Keaney Jr 1
PMCID: PMC3872835  NIHMSID: NIHMS537399  PMID: 22956414

Abstract

Cardiovascular disease continues to be a substantial health-care burden, despite recent treatment advances. Oxidative stress has long been regarded as a key pathophysiological mediator that ultimately leads to CVD including atherosclerosis, hypertension and heart failure. Over the past decade, emerging evidence has shifted our understanding of reactive oxygen species (ROS) from its harmful role to being signaling molecules. Here, we reviewed recent advances in our understanding of ROS that mediate the complex process of cardiovascular diseases, with a focus on major ROS signaling and sources such as mitochondria and NADPH oxidases.

Introduction

Our concepts of how oxidative stress contributes to chronic diseases has undergone considerable evolution in the past two decades. Historically, reactive oxygen species (ROS) and their resultant oxidative stress have been examined in the context of damage to biologically important targets such as proteins, lipids, and DNA. However, many clinical studies with antioxidants have failed to materially impact the course of human disease, including atherosclerosis and cardiovascular disease. As a consequence, the focus of how oxidative stress impacts vascular disease has shifted towards an ever increasing appreciation of oxidized targets as biomarkers and the importance of ROS as signaling molecules. With regards to the latter, research in model systems have clearly established that ROS mediate a number of physiologic and pathophysiologic processes. We are only now beginning to appreciate how these concepts are applicable to human clinical disease and this review will focus on recent clinical evidence.

Oxidative Stress: Janus-Faced Implications for Disease

With regard to cardiovascular disease, many of the pathogenic components of the disease are strongly linked to oxidative stress. For instance, LDL oxidation, endothelial dysfunction and inflammation processes are mediated, in part, by increased cellular ROS production. Similarly, other pathologic conditions associated with cardiovascular disease such as insulin resistance, metabolic syndrome, and obesity are characterized by overproduction of ROS and excess oxidative stress (1,2).

In contrast, ROS are also physiologic with one of the best examples being chronic granulomatous disease (CGD) that results from genetic defects in the phagosomal NADPH oxidase limiting ROS production (3). Patients with CGD have defective responses to pathogens manifest as recurrent infections and the lack of ROS production produces the inability to resolve innate immune responses (4). This involvement of ROS in the control of innate immunity has also been extended to mitochondrial ROS that appear to have a role in regulating Toll-like receptor responses (5). Overall, repairative processes appear to be particularly dependent upon ROS as both angiogenesis (6) and pulmonary remodeling (7).

Thus, ROS can mediate both cellular damage and physiologic events on a contextual basis. One important element in reconciling these conflicting ROS functions involves understanding the chemical nature of ROS and their molecular targets. Molecular oxygen is the parent element for ROS that are derived by sequential reduction as outlined in the scheme below:

O2+e-O2-+e-H2O2+e-OH+e-H2O

Both superoxide and hydroxyl radical are single-electron species that have limited half lives, typically act locally, and can mediate cellular damage. In contrast, H2O2 is better suited as a signaling molecule based upon its relatively longer half-life, diffusion capacity, and reaction with protein thiol moieties (8). Thus, the relative likelihood of cell damage versus signaling is dependent, in part, upon the type of ROS produced.

Another important aspect of ROS signaling versus damage is the amount of ROS produced. In the setting of inflammation, the phagocytic respiratory ROS burst produces local H2O2 concentrations between 5 and 15 μM (9). In contrast, H2O2 levels in neurons at baseline are approximately 0.2 nM, and increase to 2.5 nM with insulin treatment (10). Similarly, epidermal growth factor-induced H2O2 production yields cellular levels of 2 nM using advanced fluorescent single-walled carbon nanotube technology (11). Thus, physiological levels of H2O2 are typically less than 20 nM and cellular damage involves levels greater than 1 μM (12). This dichotomy is reminiscent of the nitric oxide system where low nM levels of NO• production by endothelial nitric oxide synthase is regulatory, whereas μM levels of NO• from inducible nitric oxide synthase contribute to the pathophysiology of septic shock. Thus, the type and amount of ROS produced provide an important basis for distinguishing physiological signaling from pathophysiological cellular damage.

ROS in Vascular Disease and Physiology

ROS and Oxidative Stress in Atherosclerosis

Atherosclerosis involves LDL entrapment in the arterial wall and an inflammatory response to the local LDL. The complexity of this process precludes a thorough discussion here. Nevertheless, common features of atherosclerosis include LDL oxidation, endothelial dysfunction, and inflammation (13). Importantly, these three features of atherosclerosis all involve ROS in their pathophysiology.

The oxidation of LDL is a well-described phenomenon in atherosclerosis. A number of enzyme systems have been proposed to contribute to LDL oxidation in vivo and there are a number of reviews available on this topic (1315). The most complete data concerning ROS-mediated LDL oxidation in vivo involve the contributions of NADPH oxidases and mitochondria. Human atherosclerotic coronary arteries contain increased immunostaining of p22phox (16), an NADPH oxidase subunit. This protein is principally associated with Nox2 in lesional macrophages, and the p22phox expression level is positively associated with atherosclerosis severity (17). Animal studies using the ApoE-null atherosclerosis model indicate that mice lacking the Nox2 isoform of NADPH oxidase exhibit a 50% reduction in lesions, along with a marked decrease in aortic ROS production, suggesting that inhibition of Nox2 NAPDPH oxidase could limit atherosclerosis (18). Humans express an NADPH oxidase isoform (Nox5) that is not found in rodents, and this oxidase may also contribute to blood vessel ROS as coronary arteries with atherosclerosis exhibit an increased expression level and activity of Nox5 (19).

Mitochondrial ROS have been implicated in many chronic diseases, including atherosclerosis (20). Emerging data now link mitochondrial ROS production to the control of inflammation. For example, mitochondrial ROS are important for signaling events critical to innate immunity (5) and activation of the NLRP3 inflammasome (21) that is known to contribute to both animal and human atherosclerosis (22).

As atherosclerotic lesions mature, they develop a fibrous cap overlying a lipid core. Acute vascular events are often caused by weakening of this fibrous cap and plaque necrosis is a key mechanism for fibrous cap weakening and rupture (23). The apoptosis of macrophages, and the inability to clear these apoptotic cells, are key contributors to plaque necrosis. When macrophages become apoptotic, they also stimulate a process known as autophagy in which the cells consume their own cellular components in an organized manner. It turns out that autophagy is an important adaptive mechanism for oxidative stress and without autophagy, ROS production (via Nox2 NADPH oxidase) is increased and plaques become more prone to rupture (24). Thus, oxidative stress is dependent upon autophagy and recent data suggests that stimulating autophagy could have beneficial effects for atherosclerosis.

ROS in Hypertension

Hypertension is an important risk factor for atherosclerosis and it also contributes to left ventricular hypertrophy and heart failure. Clinical studies have clearly indicated that inhibition of the renin-angiotensin-aldosterone system (RAS) is of major benefit for patients with hypertension (25). One potential reason for the benefit of inhibiting the RAS system is the reduction of oxidative stress and dysfunctional ROS signaling. For example, activation of the RAS system in model systems upregulates several NADPH oxidase isoforms such as Nox1 and Nox2, and subunits p22phox, p47phox, and p67phox (2628). Genetic mouse models have dissected the specific consequences of the NADPH oxidase isoforms. Animals without the Nox1 protein develop less hypertension with angiotensin II infusion (29), but exhibit the same pathologic arterial medial hypertrophy as wild-type animals. In contrast, Nox2 appears to have no impact on the blood pressure response to angiotensin II, but prevents pathologic events such as RAS-induced medial hypertrophy (30). The RAS also has implications for vascular inflammation as angiotensin infusion recruits inflammatory cells, particularly T cells, to the vascular adventitia (31). Adventitial T cells, and their ROS production, is linked to the pathophysiology of hypertension as adoptive transfer of T cells lacking functional NADPH oxidase prevents angiotensin II-induced hypertension (31). Thus, in model systems, oxidative stress in the form of NADPH oxidases plays a critical role in the pathophysiology of hypertension. These data have not yet reached full appreciation in human studies as early reports from human studies using genome-wide association strategies have yet to identify any NADPH oxidase gene loci with hypertension.

Other ROS sources have been investigated with regards to hypertension. In an animal model of salt-induced hypertension, inhibitors of the mitochondrial respiratory chain complexes I and III, as well as a mitochondrial SOD2 mimetic all demonstrated an antihypertensive effect (32), consistent with a role for mitochondria in hypertension. Similarly, antioxidants either targeted to the mitochondria (e.g. mitoTEMPO and MitoQ) or endogenously expressed in the mitochondria (SOD2 and thioredoxin 2) have all been shown to attenuate angiotensin II-induced hypertension (33,34).

One recent concept relating oxidative stress to hypertension involves cross-talk between the NADPH oxidase system and mitochondria. Endothelial NADPH oxidase-derived ROS are involved in the pathogenesis of hypertension through induction of mitochondrial dysfunction and ROS production (35). However, it also appears that mitochondria impact the NADPH oxidase system. Angiotensin treatment in animal model systems demonstrate increased mitochondrial superoxide production that results in NADPH oxidase activation as treatment with mitochondria-targeted antioxidants or excess mitochondrial SOD both limit cellular NADPH oxidase activity and angiotensin-induced hypertension (33). Thus, emerging evidence indicate both mitochondria and NADPH oxidases are important ROS sources in hypertension and may represent pathologic feed-forward mechanism. These data prompt speculation that interrupting such a vicious cycle of ROS catalyzed ROS production may hold therapeutic promise in the future.

ROS in angiogenesis

One critical vascular function is the maintenance of oxygen homoeostasis and the recruitment and development of new blood vessels in the setting of tissue ischemia. This process is dependent upon hypoxia inducible factor (HIF), a transcription factor controlling gene expression critical for angiogenesis. Mitochondria have proven critical for upregulation of HIF-1α via an ROS-dependent mechanism that involves the inhibition of prolyl hydroxylases that control HIF-1α degradation (36). Inhibition of mitochondrial ROS or mitochondria-targeted antioxidants prevent HIF-1α upregulation and angiogenesis in model systems.

Another source of ROS implicated in controlling angiogenesis include the NADPH oxidase isoforms. Mice lacking the Nox2 NADPH oxidase have defective angiogenesis responses manifest as reduced VEGF responses (37) and impaired blood flow recovery to hindlimb ischemia (38). This latter effect appears to be due to the Nox2 NADPH oxidase present in inflammatory cells rather than the vascular wall (6).

In the vascular wall, the Nox4 NADPH oxidase appears particularly important in regulating angiogenesis responses. This oxidase primarily produces H2O2 that is known to promote angiogenesis and Nox4 is upregulated in both hemangiomas (39) and ischemic hindlimb (40). Endothelial upregulation of Nox4 promotes endothelial proliferation and ROS-dependent angiogenesis (40) Conversely, mice lacking Nox4 have impaired blood flow recovery after hindlimb ischemia (41). The mechanism whereby Nox4 contributes to angiogenesis is believed to be the production of H2O2 that activates endothelial nitric oxide synthase (42) that is known to be important for angiogenesis.

Thus, from the preceding paragraphs it is clear that considerable preclinical data indicate that ROS are important modulators of repairative responses and play an important role in both the physiology and pathophysiology of vascular disease. One consequence of pathophysiologic ROS production is the modification of cellular constituents such as lipids. As a consequence, we will now turn our attention to oxidative biomarkers and their role in providing insight into the pathophysiology of vascular disease.

Oxidative Biomarker and Antioxidative Therapy

The role of ROS in cardiovascular biology and their propensity to produce oxidation byproducts has prompted considerable enthusiasm for the discovery of novel biomarkers for predicting both cardiovascular risk and therapeutic responses. Many excellent studies have characterized novel oxidation byproducts in select circumstances, however, oxidized lipoproteins, including LDL (OxLDL) have a long track record as biomarkers and appear to be among the most promising oxidation markers to potentially impact clinical practice in the near future.

Oxidized Lipids

Oxidative modification of LDL is known to be a feature of the atherosclerotic process including the initiation, progression, and destabilization of atherosclerotic plaques. Oxidized phopholipids (OxPL) are key components of OxLDL that are believed to contribute to both early and late CVD events (43). A seminal study by Tsimikas et al has demonstrated that plasma levels of OxPL present on apo B-100–containing lipoproteins and predominantly on Lp(a) lipoprotein reflect the presence and extent of angiographically documented coronary artery disease (44). Recent clinical studies have shown that elevated OxPL/apoB levels predict the risk of cardiac death, myocardial infarction and stroke, and provide cumulative predictive value when added to traditional cardiovascular risk factors (43,45). In these studies, the assay utilized a murine monoclonal antibody, E06, that specifically binds to the phosphorylcholine moiety of oxidized, but not native phospholipids. A similar strategy utilizing oxidation-specific antibody-labeled gadolinium micelles and iron oxide nanoparticles has been applied in vivo for MRI detection of atherosclerotic lesions in animals (46). This latter finding could provide a clinically useful noninvasive means of atherosclerosis imaging. Thus, oxidized phospholipids Collectively, these studies highlight the potential role of modified phospholipids as markers and/or contributors to CVD. These findings complement a recent increased interest in phospholipids in atherosclerosis, particularly since gut metabolism appears key to the mechanism of how phospholipids may contribute to CVD (47).

Myeloperoxidase

Myeloperoxidase (MPO) is a leukocyte-derived heme-containing enzyme that functions to catalyze the conversion of hydrogen peroxide (H2O2) and chloride anion (Cl) to hypochlorous acid (HOCl) as its major reaction. MPO principally is important for anti-microbial action, but is also involved in oxidation of proteins and lipids including LDL and phospholipids. Elevated circulating MPO levels are indicative of endothelial dysfunction as determined by brachial artery flow-mediated dilation, characteristic of atherosclerosis (48). In patients with acute coronary syndromes, serum MPO levels are associated with death and nonfatal MI, and this relation was independent of other biomarkers for CVD such as cardiac troponin, CRP, and soluble CD40 ligand (49). It also appears that serial MPO monitoring is able to detect a subset of at risk patients who have troponin concentrations persistently within the reference range. The results suggested that the MPO concentrations are predictive of outcome up to 16h after presentation with chest pain and predict events missed by troponin testing (50). The biologic basis for MPO participation in atherosclerosis is that MPO-modified proteins are present in human atherosclerotic lesions (51) and it is thought MPO, together with metalloproteinases, may degrade collagen within atherosclerotic plaque and result in plaque erosion or rupture (5254). Indeed, inflammatory cells producing MPO are found more frequently in ruptured plaques of patients with acute coronary syndromes than in patients with stable coronary artery disease (50,51,53,54). Thus, compelling data support the role of MPO in CAD, but it should be noted that MPO levels are influenced by numerous factors such as unfractionated heparin dosing (55) and the method of collection (56). As a consequence, more work is needed to standardize MPO as a CVD biomarker.

Other Biomarkers

There are a number of biomarkers that have showed promising results related to oxidative events in CVD. Most other markers, however, are not as mature in their development and have not been validated as extensively in multiple clinical populations compared to the oxidation markers discussed above. Thus, for the sake of brevity we have not entertained an exhaustive description of these potentially promising biomarkers.

Antioxidant Therapy

Clinical trials such as HOPE (57) and HPS (58) have not shown a benefit of Vitamin C and Vitamin E in the treatment of cardiovascular disease. The most likely explanation is that redox processes relevant to atherosclerosis and plaque rupture are not uniform and amenable to simple attenuation with antioxidants. Particularly since these antioxidants (Vitamins C and E) are already present in vivo and their effect is likely sufficient in the absence of supplementation.

Statins, although not considered antioxidants based upon their chemical structure, are a well-established treatment for atherosclerosis (59) and one of their consequences is reduced indices of oxidative stress. For example, patients treated with atorvastatin exhibit a dose-dependent decrease in circulating levels of Ox-LDL (60). Studies to investigate the mechanisms of these observations in animals demonstrated that atherosclerosis increases endothelial superoxide production and decreases NO bioavailability due to “uncoupling” of endothelial nitric oxide synthase (eNOS) whereby electrons intended for NO• production instead leak to oxygen producing superoxide. Statins have an indirect antioxidant effect by preventing eNOS uncoupling (61). An important recent study by Antoniades et al in humans indicated that atorvastatin treatment reduces vascular superoxide generation and improves vascular NO• bioavailability in human internal mammary artery of patients with atherosclerosis. The mechanism involved upregulation of GTP-cyclohydrolase I expression and activity, thereby providing complete tetrahydrobiopterin cofactor levels to improve eNOS coupling (62). Thus, statin therapy has profound implications for endothelial function.

Given that ROS production can be either physiologic or pathologic, it would be important to preferentially target antioxidants to pathologic sources of ROS production. One attempt using this strategy involves MitoQ, an ubiquinol-based-based compound that is complexed with triphenylphosphonium (TPP) cation that concentrates the molecule several hundred fold in the mitochondrion. Preclinical animal studies demonstrate that MitoQ significantly decreases heart dysfunction, cell death, and mitochondrial damage after ischemia/reperfusion (63), indicating that mitochondrial ROS are important in this condition. Similarly, previous studies had shown that mice heterozygous for the null allele of the ataxia telengectasia mutated (ATM) gene on the ApoE-null background had significant adiposity, metabolic syndrome, and atherosclerosis. Treating this model with MitoQ prevented this phenotype and also reduced the macrophage content within atherosclerotic plaques in concert with preventing oxidative DNA damage (64). Collectively, these finding suggest a potential therapeutic role for mitochondria-targeted antioxidants. Human studies to evaluate MitoQ thus far are limited, but data indicate it can be safely delivered to patients for up to a year and may be effective in decreasing liver damage in Hepatitis C patients (65). Another clinical trial with MitoQ in Parkinsons disease failed to show any effect, perhaps due to the stage of disease or limited penetration of the blood brain barrier (55,66). Nevertheless, early results suggest targeted antioxidants are a safe strategy that may hold promise in certain disease states. More work will be needed to definitively establish the role of targeted antioxidants in treating chronic disease, including atherosclerosis and CVD.

With regard to antioxidant therapy, some other factors that warrant consideration. Genetic factors are clearly involved in the etiology of CVD, thus it is plausible that the extent to which oxidative events contributes to CVD may be individualized. One must consider, therefore, that antioxidant treatment will most benefit the subset of patients who suffer from an excess of vascular oxidative stress. Thus, to properly design trials of targeted antioxidant therapy we will need to develop facile techniques to accurately assess local and systemic oxidative stress. An example of new evolving technology is the work of Albrecht et al., in which genetically encoded redox probes were used to quantitatively map both the glutathione redox potential and peroxide in defined subcellular compartments (67). Emerging technologies have expanded the scope of antioxidants to include small molecule inhibitors, nucleic acid aptamers (68), and monoclonal antibodies (69), that have the potential to target ROS in a selective manner.

Conclusions

There is a large body of evidence linking ROS to cardiovascular physiology and pathophysiology. It is now clear that the consequences of ROS are highly contextual based upon the sources and targets of these reactive molecules. As our understanding has matured, we have moved beyond simple strategies of treating patients with non-specific antioxidants to the notion that we need to target ROS modulation to specific enzyme systems and cellular compartment will inform us of the biology of ROS and potentially help risk stratification in CVD patients. With continued investigation, it is likely we will be able to target antioxidant therapy more effectively to modify the phenotypic features of cardiovascular disease.

References

Recently published papers of interest have been highlighted as

• Of importance

  • 1.Keaney JF, Larson MG, Vasan RS, Wilson PWF, Lipinska I, Corey D, Massaro JM, Sutherland P, Vita JA, Benjamin EJ Framingham Study. Obesity and systemic oxidative stress: clinical correlates of oxidative stress in the Framingham Study. Arterioscler Thromb Vasc Biol. 2003;23:434–439. doi: 10.1161/01.ATV.0000058402.34138.11. [DOI] [PubMed] [Google Scholar]
  • 2.Rao F, Zhang K, Khandrika S, Mahata M, Fung MM, Ziegler MG, Rana BK, O’Connor DT. Isoprostane, an “intermediate phenotype” for oxidative stress heritability, risk trait associations, and the influence of chromogranin B polymorphism. J Am Coll Cardiol. 2010;56:1338–1350. doi: 10.1016/j.jacc.2010.03.092. [DOI] [PubMed] [Google Scholar]
  • 3.Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 2004;4:181–189. doi: 10.1038/nri1312. [DOI] [PubMed] [Google Scholar]
  • 4.Kuijpers T, Lutter R. Inflammation and repeated infections in CGD: two sides of a coin. Cell Mol Life Sci. 2012;69:7–15. doi: 10.1007/s00018-011-0834-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, Tempst P, Walsh MC, Choi Y, Shadel GS, Ghosh S. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472:476–480. doi: 10.1038/nature09973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Urao N, Inomata H, Razvi M, Kim HW, Wary K, McKinney R, Fukai T, Ushio-Fukai M. Role of nox2-based NADPH oxidase in bone marrow and progenitor cell function involved in neovascularization induced by hindlimb ischemia. Circ Res. 2008;103:212–220. doi: 10.1161/CIRCRESAHA.108.176230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7•.Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, Martinez FJ, Thannickal VJ. NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med. 2009;15:1077–1081. doi: 10.1038/nm.2005. This article shows that TGF-beta-1-induced Nox4 expression and genetic or pharmacologic targeting of Nox4 abrogates fibrogenesis in lung injury mice models. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chen C, Thomas SR, Keaney JF. Beyond LDL oxidation: ROS in vascular signal transduction. Free Radic Biol Med. 2003;35:117–132. doi: 10.1016/s0891-5849(03)00239-9. [DOI] [PubMed] [Google Scholar]
  • 9.Liu X, Zweier JL. A real-time electrochemical technique for measurement of cellular hydrogen peroxide generation and consumption: evaluation in human polymorphonuclear leukocytes. Free Radic Biol Med. 2001;31:894–901. doi: 10.1016/s0891-5849(01)00665-7. [DOI] [PubMed] [Google Scholar]
  • 10.Storozhevykh TP, Senilova YE, Persiyantseva NA, Pinelis VG, Pomytkin IA. Mitochondrial respiratory chain is involved in insulin-stimulated hydrogen peroxide production and plays an integral role in insulin receptor autophosphorylation in neurons. BMC Neurosci. 2007;8:84. doi: 10.1186/1471-2202-8-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jin H, Heller DA, Kalbacova M, Kim J-H, Zhang J, Boghossian AA, Maheshri N, Strano MS. Detection of single-molecule H2O2 signalling from epidermal growth factor receptor using fluorescent single-walled carbon nanotubes. Nat Nanotechnol. 2010;5:302–309. doi: 10.1038/nnano.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.D’Autréaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007;8:813–824. doi: 10.1038/nrm2256. [DOI] [PubMed] [Google Scholar]
  • 13.Stocker R, Keaney JF. Role of oxidative modifications in atherosclerosis. Physiol Rev. 2004;84:1381–1478. doi: 10.1152/physrev.00047.2003. [DOI] [PubMed] [Google Scholar]
  • 14.Yoshida H, Kisugi R. Mechanisms of LDL oxidation. Clin Chim Acta. 2010;411:1875–1882. doi: 10.1016/j.cca.2010.08.038. [DOI] [PubMed] [Google Scholar]
  • 15.Levitan I, Volkov S, Subbaiah PV. Oxidized LDL: diversity, patterns of recognition, and pathophysiology. Antioxid Redox Signal. 2010;13:39–75. doi: 10.1089/ars.2009.2733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Azumi H, Inoue N, Takeshita S, Rikitake Y, Kawashima S, Hayashi Y, Itoh H, Yokoyama M. Expression of NADH/NADPH oxidase p22phox in human coronary arteries. Circulation. 1999;100:1494–1498. doi: 10.1161/01.cir.100.14.1494. [DOI] [PubMed] [Google Scholar]
  • 17.Sorescu D, Weiss D, Lassègue B, Clempus RE, Szöcs K, Sorescu GP, Valppu L, Quinn MT, Lambeth JD, Vega JD, Taylor WR, Griendling KK. Superoxide production and expression of nox family proteins in human atherosclerosis. Circulation. 2002;105:1429–1435. doi: 10.1161/01.cir.0000012917.74432.66. [DOI] [PubMed] [Google Scholar]
  • 18.Judkins CP, Diep H, Broughton BRS, Mast AE, Hooker EU, Miller AA, Selemidis S, Dusting GJ, Sobey CG, Drummond GR. Direct evidence of a role for Nox2 in superoxide production, reduced nitric oxide bioavailability, and early atherosclerotic plaque formation in ApoE−/− mice. Am J Physiol Heart Circ Physiol. 2010;298:H24–32. doi: 10.1152/ajpheart.00799.2009. [DOI] [PubMed] [Google Scholar]
  • 19.Guzik TJ, Chen W, Gongora MC, Guzik B, Lob HE, Mangalat D, Hoch N, Dikalov S, Rudzinski P, Kapelak B, Sadowski J, Harrison DG. Calcium-dependent NOX5 nicotinamide adenine dinucleotide phosphate oxidase contributes to vascular oxidative stress in human coronary artery disease. J Am Coll Cardiol. 2008;52:1803–1809. doi: 10.1016/j.jacc.2008.07.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Corral-Debrinski M, Shoffner JM, Lott MT, Wallace DC. Association of mitochondrial DNA damage with aging and coronary atherosclerotic heart disease. Mutat Res. 1992;275:169–180. doi: 10.1016/0921-8734(92)90021-g. [DOI] [PubMed] [Google Scholar]
  • 21.Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:221–225. doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
  • 22.Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361. doi: 10.1038/nature08938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2000;20:1262–1275. doi: 10.1161/01.atv.20.5.1262. [DOI] [PubMed] [Google Scholar]
  • 24•.Liao X, Sluimer JC, Wang Y, Subramanian M, Brown K, Pattison JS, Robbins J, Martinez J, Tabas I. Macrophage autophagy plays a protective role in advanced atherosclerosis. Cell Metab. 2012;15:545–553. doi: 10.1016/j.cmet.2012.01.022. This article showed that inhibition of autophagy promotes apoptosis and NADPH oxidase-mediated oxidative stress, and defective efferocytosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Epstein BJ, Smith SM, Choksi R. Recent changes in the landscape of combination RAS blockade. Expert Rev Cardiovasc Ther. 2009;7:1373–1384. doi: 10.1586/erc.09.127. [DOI] [PubMed] [Google Scholar]
  • 26.Chabrashvili T, Kitiyakara C, Blau J, Karber A, Aslam S, Welch WJ, Wilcox CS. Effects of ANG II type 1 and 2 receptors on oxidative stress, renal NADPH oxidase, and SOD expression. Am J Physiol Regul Integr Comp Physiol. 2003;285:R117–24. doi: 10.1152/ajpregu.00476.2002. [DOI] [PubMed] [Google Scholar]
  • 27.Kitiyakara C, Chabrashvili T, Chen Y, Blau J, Karber A, Aslam S, Welch WJ, Wilcox CS. Salt intake, oxidative stress, and renal expression of NADPH oxidase and superoxide dismutase. J Am Soc Nephrol. 2003;14:2775–2782. doi: 10.1097/01.asn.0000092145.90389.65. [DOI] [PubMed] [Google Scholar]
  • 28•.Feng D, Yang C, Geurts AM, Kurth T, Liang M, Lazar J, Mattson DL, O’Connor PM, Cowley AW. Increased expression of NAD(P)H oxidase subunit p67(phox) in the renal medulla contributes to excess oxidative stress and salt-sensitive hypertension. Cell Metab. 2012;15:201–208. doi: 10.1016/j.cmet.2012.01.003. This article shows that the higher expression of p67(phox), not the other subunits, was associated with higher Nox activity and salt sensitivity, and disruption of p67(phox) resulted in reduction of salt-sensitive hypertension and renal medullary oxidative stress/injury. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Matsuno K, Yamada H, Iwata K, Jin D, Katsuyama M, Matsuki M, Takai S, Yamanishi K, Miyazaki M, Matsubara H, Yabe-Nishimura C. Nox1 is involved in angiotensin II-mediated hypertension: a study in Nox1-deficient mice. Circulation. 2005;112:2677–2685. doi: 10.1161/CIRCULATIONAHA.105.573709. [DOI] [PubMed] [Google Scholar]
  • 30.Wang HD, Xu S, Johns DG, Du Y, Quinn MT, Cayatte AJ, Cohen RA. Role of NADPH oxidase in the vascular hypertrophic and oxidative stress response to angiotensin II in mice. Circ Res. 2001;88:947–953. doi: 10.1161/hh0901.089987. [DOI] [PubMed] [Google Scholar]
  • 31.Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007;204:2449–2460. doi: 10.1084/jem.20070657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang A, Jia Z, Wang N, Tidwell TJ, Yang T. Relative contributions of mitochondria and NADPH oxidase to deoxycorticosterone acetate-salt hypertension in mice. Kidney Int. 2011;80:51–60. doi: 10.1038/ki.2011.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33•.Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W, Harrison DG, Dikalov SI. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res. 2010;107:106–116. doi: 10.1161/CIRCRESAHA.109.214601. This article shows that treatment with the mitochondrial-targeted antioxidant mitoTEMPO attenuated hypertension and overexpression of SOD2 in transgenic mice demonstreated attenuated angiotensin-II-induced hypertension. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Widder JD, Fraccarollo D, Galuppo P, Hansen JM, Jones DP, Ertl G, Bauersachs J. Attenuation of angiotensin II-induced vascular dysfunction and hypertension by overexpression of Thioredoxin 2. Hypertension. 2009;54:338–344. doi: 10.1161/HYPERTENSIONAHA.108.127928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Doughan AK, Harrison DG, Dikalov SI. Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunction. Circ Res. 2008;102:488–496. doi: 10.1161/CIRCRESAHA.107.162800. [DOI] [PubMed] [Google Scholar]
  • 36.Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell. 2012;148:399–408. doi: 10.1016/j.cell.2012.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ushio-Fukai M, Tang Y, Fukai T, Dikalov SI, Ma Y, Fujimoto M, Quinn MT, Pagano PJ, Johnson C, Alexander RW. Novel role of gp91(phox)-containing NAD(P)H oxidase in vascular endothelial growth factor-induced signaling and angiogenesis. Circ Res. 2002;91:1160–1167. doi: 10.1161/01.res.0000046227.65158.f8. [DOI] [PubMed] [Google Scholar]
  • 38.Tojo T, Ushio-Fukai M, Yamaoka-Tojo M, Ikeda S, Patrushev N, Alexander RW. Role of gp91phox (Nox2)-containing NAD(P)H oxidase in angiogenesis in response to hindlimb ischemia. Circulation. 2005;111:2347–2355. doi: 10.1161/01.CIR.0000164261.62586.14. [DOI] [PubMed] [Google Scholar]
  • 39.Bhandarkar SS, Jaconi M, Fried LE, Bonner MY, Lefkove B, Govindarajan B, Perry BN, Parhar R, Mackelfresh J, Sohn A, Stouffs M, Knaus U, Yancopoulos G, Reiss Y, Benest AV, Augustin HG, Arbiser JL. Fulvene-5 potently inhibits NADPH oxidase 4 and blocks the growth of endothelial tumors in mice. J Clin Invest. 2009;119:2359–2365. doi: 10.1172/JCI33877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40•.Craige SM, Chen C, Pei Y, Li C, Huang X, Chen C, Shibata R, Sato K, Walsh K, Keaney JF. NADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation. Circulation. 2011;124:731–740. doi: 10.1161/CIRCULATIONAHA.111.030775. This article shows endothelial-specific Nox4-overexpressing mice had accelerated recovery from hindlimb ischemia through enhanced angiogenesis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41•.Schröder K, Zhang M, Benkhoff S, Mieth A, Pliquett R, Kosowski J, Kruse C, Lüdike P, Michaelis UR, Weissmann N, Dimmeler S, Shah AM, Brandes RP. Nox4 Is a Protective Reactive Oxygen Species Generating Vascular NADPH Oxidase. Circ Res. 2012 doi: 10.1161/CIRCRESAHA.112.267054. in press. This article shows that Nox4-null mice exhibited attenuated angiogenesis and reduced eNOS expression. [DOI] [PubMed] [Google Scholar]
  • 42.Thomas SR, Chen C, Keaney JF. Hydrogen peroxide activates endothelial nitric-oxide synthase through coordinated phosphorylation and dephosphorylation via a phosphoinositide 3-kinase-dependent signaling pathway. J Biol Chem. 2002;277:6017–6024. doi: 10.1074/jbc.M109107200. [DOI] [PubMed] [Google Scholar]
  • 43.Hartvigsen K, Chou M-Y, Hansen LF, Shaw PX, Tsimikas S, Binder CJ, Witztum JL. The role of innate immunity in atherogenesis. J Lipid Res. 2009;50 (Suppl):S388–93. doi: 10.1194/jlr.R800100-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tsimikas S, Brilakis ES, Miller ER, McConnell JP, Lennon RJ, Kornman KS, Witztum JL, Berger PB. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N Engl J Med. 2005;353:46–57. doi: 10.1056/NEJMoa043175. [DOI] [PubMed] [Google Scholar]
  • 45.Kiechl S, Willeit J, Mayr M, Viehweider B, Oberhollenzer M, Kronenberg F, Wiedermann CJ, Oberthaler S, Xu Q, Witztum JL, Tsimikas S. Oxidized phospholipids, lipoprotein(a), lipoprotein-associated phospholipase A2 activity, and 10-year cardiovascular outcomes: prospective results from the Bruneck study. Arterioscler Thromb Vasc Biol. 2007;27:1788–1795. doi: 10.1161/ATVBAHA.107.145805. [DOI] [PubMed] [Google Scholar]
  • 46•.Briley-Saebo KC, Cho Y-S, Shaw PX, Ryu SK, Mani V, Dickson S, Izadmehr E, Green S, Fayad ZA, Tsimikas S. Targeted iron oxide particles for in vivo magnetic resonance detection of atherosclerotic lesions with antibodies directed to oxidation-specific epitopes. J Am Coll Cardiol. 2011;57:337–347. doi: 10.1016/j.jacc.2010.09.023. This article shows following intravenous injection of targeted Mn micelles, strong MR signal was observed 48–72 h with colocalization within intraplaque macrophages. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung Y-M, Wu Y, Schauer P, Smith JD, Allayee H, Tang WHW, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Vita JA, Brennan M-L, Gokce N, Mann SA, Goormastic M, Shishehbor MH, Penn MS, Keaney JF, Hazen SL. Serum myeloperoxidase levels independently predict endothelial dysfunction in humans. Circulation. 2004;110:1134–1139. doi: 10.1161/01.CIR.0000140262.20831.8F. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Baldus S, Heeschen C, Meinertz T, Zeiher AM, Eiserich JP, Münzel T, Simoons ML, Hamm CW CAPTURE Investigators. Myeloperoxidase serum levels predict risk in patients with acute coronary syndromes. Circulation. 2003;108:1440–1445. doi: 10.1161/01.CIR.0000090690.67322.51. [DOI] [PubMed] [Google Scholar]
  • 50.Nicholls SJ, Tang WHW, Brennan D, Brennan M-L, Mann S, Nissen SE, Hazen SL. Risk prediction with serial myeloperoxidase monitoring in patients with acute chest pain. Clin Chem. 2011;57:1762–1770. doi: 10.1373/clinchem.2011.166827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hazell LJ, Arnold L, Flowers D, Waeg G, Malle E, Stocker R. Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest. 1996;97:1535–1544. doi: 10.1172/JCI118576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sugiyama S, Okada Y, Sukhova GK, Virmani R, Heinecke JW, Libby P. Macrophage myeloperoxidase regulation by granulocyte macrophage colony-stimulating factor in human atherosclerosis and implications in acute coronary syndromes. Am J Pathol. 2001;158:879–891. doi: 10.1016/S0002-9440(10)64036-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Naruko T, Ueda M, Haze K, van der Wal AC, van der Loos CM, Itoh A, Komatsu R, Ikura Y, Ogami M, Shimada Y, Ehara S, Yoshiyama M, Takeuchi K, Yoshikawa J, Becker AE. Neutrophil infiltration of culprit lesions in acute coronary syndromes. Circulation. 2002;106:2894–2900. doi: 10.1161/01.cir.0000042674.89762.20. [DOI] [PubMed] [Google Scholar]
  • 54.Buffon A, Biasucci LM, Liuzzo G, D’Onofrio G, Crea F, Maseri A. Widespread coronary inflammation in unstable angina. N Engl J Med. 2002;347:5–12. doi: 10.1056/NEJMoa012295. [DOI] [PubMed] [Google Scholar]
  • 55.Baldus S, Rudolph V, Roiss M, Ito WD, Rudolph TK, Eiserich JP, Sydow K, Lau D, Szöcs K, Klinke A, Kubala L, Berglund L, Schrepfer S, Deuse T, Haddad M, Risius T, Klemm H, Reichenspurner HC, Meinertz T, Heitzer T. Heparins increase endothelial nitric oxide bioavailability by liberating vessel-immobilized myeloperoxidase. Circulation. 2006;113:1871–1878. doi: 10.1161/CIRCULATIONAHA.105.590083. [DOI] [PubMed] [Google Scholar]
  • 56.Shih J, Datwyler SA, Hsu SC, Matias MS, Pacenti DP, Lueders C, Mueller C, Danne O, Möckel M. Effect of collection tube type and preanalytical handling on myeloperoxidase concentrations. Clin Chem. 2008;54:1076–1079. doi: 10.1373/clinchem.2007.101568. [DOI] [PubMed] [Google Scholar]
  • 57.McQueen MJ, Lonn E, Gerstein HC, Bosch J, Yusuf S. The HOPE (Heart Outcomes Prevention Evaluation) Study and its consequences. Scand J Clin Lab Invest Suppl. 2005;240:143–156. doi: 10.1080/00365510500236366. [DOI] [PubMed] [Google Scholar]
  • 58.Bates CJ. MRC/BHF Heart Protection Study. Lancet. 2002;360:1781–2. doi: 10.1016/s0140-6736(02)11689-8. author reply 1783–4. [DOI] [PubMed] [Google Scholar]
  • 59.Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, Macfadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ JUPITER Trial Study Group. Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009;373:1175–1182. doi: 10.1016/S0140-6736(09)60447-5. [DOI] [PubMed] [Google Scholar]
  • 60.Singh U, Devaraj S, Jialal I, Siegel D. Comparison effect of atorvastatin (10 versus 80 mg) on biomarkers of inflammation and oxidative stress in subjects with metabolic syndrome. Am J Cardiol. 2008;102:321–325. doi: 10.1016/j.amjcard.2008.03.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang C-Y, Liu P-Y, Liao JK. Pleiotropic effects of statin therapy: molecular mechanisms and clinical results. Trends Mol Med. 2008;14:37–44. doi: 10.1016/j.molmed.2007.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62•.Antoniades C, Bakogiannis C, Leeson P, Guzik TJ, Zhang M-H, Tousoulis D, Antonopoulos AS, Demosthenous M, Marinou K, Hale A, Paschalis A, Psarros C, Triantafyllou C, Bendall J, Casadei B, Stefanadis C, Channon KM. Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling. Circulation. 2011;124:335–345. doi: 10.1161/CIRCULATIONAHA.110.985150. This article shows statin treatment in patients undergoing coronary artery bypass graft surgery was associated with improved vascular nitric oxide bioavailability and reduced superoxide generation in internal mammary artery. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Adlam VJ, Harrison JC, Porteous CM, James AM, Smith RAJ, Murphy MP, Sammut IA. Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. FASEB J. 2005;19:1088–1095. doi: 10.1096/fj.05-3718com. [DOI] [PubMed] [Google Scholar]
  • 64•.Mercer JR, Yu E, Figg N, Cheng K-K, Prime TA, Griffin JL, Masoodi M, Vidal-Puig A, Murphy MP, Bennett MR. The mitochondria-targeted antioxidant MitoQ decreases features of the metabolic syndrome in ATM+/−/ApoE−/− mice. Free Radic Biol Med. 2012;52:841–849. doi: 10.1016/j.freeradbiomed.2011.11.026. This article shows that MitoQ intake orally prevented the increased adposity, hypercholesterolemia, and hypertriglyceridemia, and corrected hyperglycemia and hepatic steatosis. [DOI] [PubMed] [Google Scholar]
  • 65•.Gane EJ, Weilert F, Orr DW, Keogh GF, Gibson M, Lockhart MM, Frampton CM, Taylor KM, Smith RAJ, Murphy MP. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int. 2010;30:1019–1026. doi: 10.1111/j.1478-3231.2010.02250.x. This article shows that treatment with oral MitoQ in patients with hepatitis C decreased alanine transaminase and aspartate aminotransferase levels without changes in viral load. [DOI] [PubMed] [Google Scholar]
  • 66.Smith RAJ, Murphy MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci. 2010;1201:96–103. doi: 10.1111/j.1749-6632.2010.05627.x. [DOI] [PubMed] [Google Scholar]
  • 67.Albrecht SC, Barata AG, Grosshans J, Teleman AA, Dick TP. In vivo mapping of hydrogen peroxide and oxidized glutathione reveals chemical and regional specificity of redox homeostasis. Cell Metab. 2011;14:819–829. doi: 10.1016/j.cmet.2011.10.010. [DOI] [PubMed] [Google Scholar]
  • 68.Keefe AD, Pai S, Ellington A. Aptamers as therapeutics. Nat Rev Drug Discov. 2010;9:537–550. doi: 10.1038/nrd3141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhang L, Nguyen MVC, Lardy B, Jesaitis AJ, Grichine A, Rousset F, Talbot M, Paclet M-H, Qian G, Morel F. New insight into the Nox4 subcellular localization in HEK293 cells: first monoclonal antibodies against Nox4. Biochimie. 2011;93:457–468. doi: 10.1016/j.biochi.2010.11.001. [DOI] [PubMed] [Google Scholar]

RESOURCES