Abstract

HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 24, No. 7Myeloperoxidase and Plaque Vulnerability Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBMyeloperoxidase and Plaque Vulnerability Stanley L. Hazen Stanley L. HazenStanley L. Hazen From the Departments of Cell Biology, Cardiovascular Medicine, and the Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic Foundation, Cleveland, Ohio 44195 Search for more papers by this author Originally published1 Jul 2004https://doi.org/10.1161/01.ATV.0000135267.82813.52Arteriosclerosis, Thrombosis, and Vascular Biology. 2004;24:1143–1146Inflammation and the Vulnerable PlaqueSudden cardiac death remains the leading cause of mortality in industrialized societies, outpacing all cancer related deaths combined. The majority of sudden cardiac deaths arise from acute myocardial infarction secondary to intracoronary artery thromboses. Remarkably, the culprit lesions involved are typically not flow-limiting stenoses,1,2 but rather inflamed lipid-laden lesions.3,4 Whereas plaque fissuring or rupture, which exposes the intensely prothrombogenic lipid core, occurs in a majority of cases, fully 40% of intracoronary artery thromboses arise at sites of superficial erosions, where endothelial cell (EC) loss and denudation occurs.3 The mechanisms responsible for plaque vulnerability leading to acute coronary artery thrombosis remain poorly understood. Mounting evidence, however, points toward a critical role for inflammatory processes.5–8 Macrophages serve as the dominant cell type in the immediate site of both plaque ruptures and superficial erosions in subjects who experience acute coronary thrombosis,8 and recent clinical investigations reveal important associations between leukocytes,9,10 their enzymes,11–13 and their activation,14,15 in subjects with unstable angina and acute coronary syndromes. In this issue of Arteriosclerosis, Thrombosis, and Vascular Biology, Sugiyama and colleagues significantly extend our knowledge of potential pathophysiologic inflammatory processes within vulnerable atheroma.16 Using a combination of biochemical, cellular, and immunohistologic studies, they describe unifying mechanistic links between the activity of the leukocyte enzyme myeloperoxidase (MPO) and two cardinal features of vulnerable plaque: EC loss/denudation and development of a prothrombotic phenotype.See page 1309Myeloperoxidase: an Inflammatory Mediator of AtherosclerosisFirst identified within human atherosclerotic plaque nearly a decade ago,17 MPO has emerged as an important potential participant in the atherosclerotic process. MPO, a member of the heme peroxidase superfamily, generates reactive oxidants and diffusible radical species as part of its normal function in innate host defenses.18 A unique activity of MPO is its ability to use the halide chloride as cosubstrate with hydrogen peroxide to generate chlorinating oxidants such as hypochlorous acid (HOCl), a potent antimicrobicidal agent.18–20 MPO,17,21 and specific chlorinated protein20 and lipid22 oxidation products, are all markedly enriched within human atheroma. Leukocytes use MPO to generate oxidants capable of initiating lipid peroxidation both in model systems23,24 and in vivo,25 including conversion of low density lipoprotein into an atherogenic form recognized by macrophage scavenger receptors.26 MPO may also contribute to the atherosclerotic process by promoting endothelial dysfunction, by virtue of its capacity to catalytically consume nitric oxide as a substrate in vitro27 and in vivo,28 resulting in formation of nitric oxide–derived oxidants.29–31 Indeed, recent clinical studies demonstrate that systemic levels of MPO serve as a strong and independent predictor of endothelial dysfunction in subjects,32 as well as angiographic evidence of CAD.11 Finally, recent human genetic studies support a potential role for MPO in CAD because MPO deficiency in subjects is reportedly cardioprotective,33 and individuals possessing a functional polymorphism associated with approximately two-fold decrease in MPO expression have reduced cardiac risks.34–36Myeloperoxidase and Plaque VulnerabilityDespite the many links between MPO and CAD development and progression, only recently has a potential role for MPO and its oxidants in development of vulnerable plaque been addressed. Libby and colleagues first described the strong colocalization between macrophage MPO expression and HOCl-modified proteins within culprit lesions of subjects with sudden cardiac death, suggesting a potential mechanistic role for MPO.21 Subsequent clinical studies by Brennan et al12 and Baldus and colleagues13 both revealed the potential utility of circulating MPO levels as a predictor of plaque vulnerability in subjects at risk for incident major adverse cardiac events, even in the absence of detectable levels of myocardial necrosis. Although the ability of MPO to activate latent matrix metalloproteinases into active forms37 may play a role in the association of MPO immunostaining at sites of plaque fissuring, potential mechanisms linking MPO to development of superficial coronary erosions and a prothrombotic endothelium remained essentially unexplored.In this issue, Sugiyama and colleagues16 extend these prior observations by demonstrating that MPO-generated HOCl, at doses likely formed at sites of inflamed vascular lesions, provokes a biphasic response in human ECs. Low doses of HOCl (<10 μmol/L) led to EC activation and elaboration of tissue factor messenger RNA, protein, and tissue factor pathway activity (Figure). Higher yet physiologically relevant doses (30 to 50 μmol/L) promoted EC death and detachment by apparent apoptotic mechanisms, based on demonstrations of rapid caspace-3 activation, decreased EC Bcl-2, cytochrome-C release, poly (ADP-ribose) polymerase degradation, and DNA laddering. Similar phenomena were observed using MPO-positive, but not MPO-negative, human macrophages, suggesting that MPO-generated HOCl within the subendothelium may contribute to plaque vulnerability by evoking EC death, plaque erosion, and induction of a prothrombogenic surface (Figure). A critical role for depletion of intracellular reduced glutathione (GSH), a kinetically favored target of HOCl oxidation,38 was suggested in their studies by demonstrating reversal of HOCl-, MPO-, and MPO-positive macrophage-mediated effects by either preloading EC with GSH esters or pretreatment with the cell-permeable statin cerivastatin. Download figureDownload PowerPointMechanisms linking myeloperoxidase to characteristic features of vulnerable plaque (endothelial cell loss/denudation and a prothrombotic phenotype). Low levels of MPO-generated HOCl activate endothelial cells, leading to tissue factor expression and activation. Higher levels of the MPO-generated oxidant promote endothelial cell apoptosis. Previous studies demonstrate the colocalization of both MPO and HOCl-modified proteins in subendothelium of culprit lesions of patients with sudden cardiac death, including enrichment at sites of both plaque fissuring or rupture, and superficial erosions.21An attractive feature of Sugiyama et al’s study is that it provides a unifying mechanistic framework accounting for numerous clinical, histological, biochemical, and cellular results linking MPO and oxidant stress with EC apoptosis, tissue factor pathway activity, and development of vulnerable plaque. Loss of endothelial functional integrity with accompanying endothelial dysfunction appears to be a common molecular disorder of unstable atherosclerotic vascular disease.39–42 There is a growing appreciation of the prevalence of EC apoptosis in atheroma, and increased levels of apoptotic ECs are reported within the systemic circulation of subjects with symptomatic atherosclerotic disease.43,44 The demonstration that MPO-containing macrophages provoke EC apoptosis16 may serve as a mechanism contributing to the previously observed colocalization of both MPO and HOCl-modified proteins within culprit lesions of subjects with sudden cardiac death.21 Similarly, demonstration of EC tissue factor expression and activation in response to physiological levels of HOCl16 provides a mechanistic rationale for the previously reported association between MPO and HOCl-modified proteins at sites of intracoronary artery thromboses in autopsy specimens.16 An additional potential mechanism for the association may include the demonstration that lipid hydroperoxides, species generated by MPO in vivo,25 promote the activation of latent tissue factor pathway activity.45,46 Additionally, MPO-generated oxidation products of plasmalogens were recently shown to be both markedly enriched within human atheroma and capable of promoting EC activation and P-selectin surface expression.22Vermani and colleagues reported the observation that superficial coronary artery erosions with accompanying occlusive intraarterial thrombi occur in more than a quarter of cases of sudden cardiac death.3,42 Remarkably, superficial erosions, as opposed to a fissure or crack within the fibrous plaque, are observed three times as often in females compared with males.3,4 The mechanism(s) responsible for this female gender-preference is unknown. Alterations in matrix via protease activation likely contribute to plaque destabilization and rupture, whereas a primary role for EC injury and desquamation seems probable in superficial erosions. Given the reported findings of Sugiyama et al in this issue of the Journal, and their prior report that sites of superficial coronary erosions possess subendothelium enriched in MPO and HOCl-modified proteins,21 one might speculate that MPO-generated HOCl-mediated EC apoptosis may partly underlie the female sex-specific difference. To further explore the possibility that MPO differentially affects cardiovascular risks based on sex, we felt it would be informative to perform sex-specific analyses of our recently reported clinical study exploring the predictive value of plasma MPO levels in subjects with vulnerable plaque.12 Of interest, there is a suggestion that MPO levels may demonstrate sex-specific differences in cardiovascular risks (Table). Although plasma levels of MPO tended to be lower in females (P=0.05), they showed a tendency toward being a stronger predictor of risk in females than in males (P=0.08) (Table). It is of interest to note that estradiol has recently been identified as a potential endogenous substrate for MPO in plasma that is capable of initiating lipid peroxidation.24 Whether these findings are linked and explain the female predilection for coronary erosions with underlying MPO-laden plaque remains unknown. Further evaluation into this area is warranted. Gender Associated Effects on Myeloperoxidase Levels and Odds Ratio for Major Adverse Cardiac Events (30 days)Age (y)MPO (pM)RR (95%CI)Q2Q3Q4119.4–197.9198.0–393.9>394.0Examination of plasma MPO levels in 604 sequential subjects (250 female) presenting to the emergency room with chest pain suggests that MPO may serve as a stronger predictor of incident major adverse cardiac events (MI, need for revascularization, and cardiac death) in the ensuing 30 days interval in females compared to males (P=0.08). Values for age and MPO levels are reported as median and interquartile ranges. Relative risks (RR) and 95% confidence intervals for each MPO quartile (Q) are shown. Adapted from clinical data described within Brennan et al12.Male63.9 (51.2–72.7)192.1 (112.5–376.4)1.4 (0.7–2.9)2.5 (1.3–4.7)3.5 (1.9–6.5)Female64.8 (51.5–75.1)166.2 (97.3–334.6)2.5 (1.0–6.3)4.7 (1.9–11.7)8.3 (3.4–20.2)In summary, a growing body of evidence suggests important mechanistic links between inflammation and development of the vulnerable plaque phenotype. MPO is emerging as a potential prognostic indicator of near term cardiovascular risks, as well as a participant in the underlying pathophysiological processes. Development of an MPO inhibitor may represent a novel therapeutic strategy for preventing or interrupting development of vulnerable plaque.This work was supported by grants from the National Institutes of HealthFootnotesCorrespondence to Stanley L. Hazen, MD, PhD, Preventive Cardiology, Cleveland Clinic Foundation, 9500 Euclid Avenue, C51, Cleveland, OH 44195. E-mail [email protected] References 1 Qiao JH, Fishbein MC. The severity of coronary atherosclerosis at sites of plaque rupture with occlusive thrombosis. J Am Coll Cardiol. 1991; 17: 1138–1142.CrossrefMedlineGoogle Scholar2 Giroud D, Li JM, Urban P, Meier B, Rutishauer W. Relation of the site of acute myocardial infarction to the most severe coronary arterial stenosis at prior angiography. Am J Cardiol. 1992; 69: 729–732.CrossrefMedlineGoogle Scholar3 Farb A, Burke AP, Tang AL, Liang TY, Mannan P, Smialek J, Virmani R. Coronary plaque erosion without rupture into a lipid core. A frequent cause of coronary thrombosis in sudden coronary death. Circulation. 1996; 93: 1354–1363.CrossrefMedlineGoogle Scholar4 Arbustini E, Dal Bello B, Morbini P, Burke AP, Bocciarelli M, Specchia G, Virmani R. Plaque erosion is a major substrate for coronary thrombosis in acute myocardial infarction. Heart. 1999; 82: 269–272.CrossrefMedlineGoogle Scholar5 Lusis AJ. Atherosclerosis. Nature. 2000; 407 (6801): 233–241.CrossrefMedlineGoogle Scholar6 Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation. 2002; 105: 1135–1143.CrossrefMedlineGoogle Scholar7 Robbins M, Topol EJ. Inflammation in acute coronary syndromes. Cleve Clin J Med. 2002; 69 (Suppl 2): SII130–SII42.MedlineGoogle Scholar8 Burke AP, Farb A, Malcom GT, Liang YH, Smialek J, Virmani R. Coronary risk factors and plaque morphology in men with coronary disease who died suddenly. N Engl J Med. 1997; 336: 1276–1282.CrossrefMedlineGoogle Scholar9 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.LinkGoogle Scholar10 Gurm HS, Bhatt DL, Gupta R, Ellis SG, Topol EJ, Lauer MS. Preprocedural white blood cell count and death after percutaneous coronary intervention. Am Heart J. 2003; 146: 692–698.CrossrefMedlineGoogle Scholar11 Zhang R, Brennan ML, Fu X, Aviles RJ, Pearce GL, Penn MS, Topol EJ, Sprecher DL, Hazen SL. Association between myeloperoxidase levels and risk of coronary artery disease. JAMA. 2001; 286: 2136–2142.CrossrefMedlineGoogle Scholar12 Brennan ML, Penn MS, Van Lente F, Nambi V, Shishehbor MH, Aviles RJ, Goormastic M, Pepoy ML, McErlean ES, Topol EJ, Nissen SE, Hazen SL. Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med. 2003; 349: 1595–1604.CrossrefMedlineGoogle Scholar13 Baldus S, Heeschen C, Meinertz T, Zeiher AM, Eiserich JP, Munzel T, Simoons ML, Hamm CW. Myeloperoxidase serum levels predict risk in patients with acute coronary syndromes. Circulation. 2003; 108: 1440–1445.LinkGoogle Scholar14 Takeshita S, Hashimoto H, Ono Y, Ochiai M, Yokoyama N, Terakura M, Sato T, Isshiki T. Increased leukocyte activity as a predictor for flow-limiting coronary lesions in patients with angina pectoris. Atherosclerosis. 2001; 158: 477–481.CrossrefMedlineGoogle Scholar15 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.CrossrefMedlineGoogle Scholar16 Sugiyama S, Kugiyama K, Aikawa M, Nakamura S, Ogawa H, Libby P. Hypochlorous Acid, a Macrophage Product, Induces Endothelial Apoptosis and Tissue Factor Expression. Involvement of Myeloperoxidase-Mediated Oxidant in Plaque Erosion and Thrombogenesis. Arterioscler Thromb Vasc Biol. 2004;1309–1314.MedlineGoogle Scholar17 Daugherty A, Dunn JL, Rateri DL, Heinecke JW. Myeloperoxidase, a catalyst for lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest. 1994; 94: 437–444.CrossrefMedlineGoogle Scholar18 Klebanoff SJ. Oxygen metabolism and the toxic properties of phagocytes. Ann Intern Med. 1980; 93: 480–489.CrossrefMedlineGoogle Scholar19 Hurst JK, Barrette WC Jr. Leukocytic oxygen activation and microbicidal oxidative toxins. Crit Rev Biochem Mol Biol. 1989; 24: 271–328.CrossrefMedlineGoogle Scholar20 Hazen SL, Heinecke JW. 3-Chlorotyrosine, a specific marker of myeloperoxidase-catalyzed oxidation, is markedly elevated in low density lipoprotein isolated from human atherosclerotic intima. J Clin Invest. 1997; 99: 2075–2081.CrossrefMedlineGoogle Scholar21 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.CrossrefMedlineGoogle Scholar22 Thukkani AK, McHowat J, Hsu FF, Brennan ML, Hazen SL, Ford DA. Identification of alpha-chloro fatty aldehydes and unsaturated lysophosphatidylcholine molecular species in human atherosclerotic lesions. Circulation. 2003; 108: 3128–3133.LinkGoogle Scholar23 Hazen SL, Zhang R, Wu W, Podrez EA, Shen Z, MacPhearson J, Schmitt D, Mitra SN, Chen Y, Cohen P, Hoff HF, Abu-Soud HM. Formation of nitric oxide–derived oxidants by myeloperoxidase in monocytes: pathways for monocyte-mediated protein nitration and lipid peroxidation in vivo. Circ Res. 1999; 85: 950–958.CrossrefMedlineGoogle Scholar24 Zhang R, Shen Z, Nauseef WM, Hazen SL. The role of myeloperoxidase in the initiation of lipid peroxidation in plasma as studied in neutrophils isolated from myeloperoxidase deficient subjects: Systematic approach to the identification and characterization of multiple diffusible endogenous substrates for myeloperoxidase in plasma. Blood. 2002; 99: 1802–1810.CrossrefMedlineGoogle Scholar25 Zhang R, Brennan ML, Shen Z, MacPherson JC, Molenda CE, Hazen SL. Myeloperoxidase functions as a major enzymatic catalyst for initiation of lipid peroxidation at sites of inflammation. J Biol Chem. 2002; 277: 46116–46122.CrossrefMedlineGoogle Scholar26 Podrez EA, Febbraio M, Sheibani N, Schmitt D, Silverstein R, Hajjar DP, Cohen PA, Frazier WA, Hoff HF, Hazen SL. The macrophage scavenger receptor CD36 is the major receptor for LDL recognition following modification by monocyte-generated reactive nitrogen species. J Clin Invest. 2000; 105: 1095–1108.CrossrefMedlineGoogle Scholar27 Abu-Soud HM, Hazen SL. Nitric oxide is a physiological substrate for mammalian peroxidases. J Biol Chem. 2000; 275: 37524–37532.CrossrefMedlineGoogle Scholar28 Eiserich JP, Baldus S, Brennan ML, Ma W, Zhang C, Tousson A, Castro L, Lusis AJ, Nauseef WM, White CR, Freeman BA. Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Science. 2002; 296 (5577): 2391–2394.CrossrefMedlineGoogle Scholar29 Baldus S, Eiserich JP, Mani A, Castro L, Figueroa M, Chumley P, Ma W, Tousson A, White CR, Bullard DC, Brennan ML, Lusis AJ, Moore KP, Freeman BA. Endothelial transcytosis of myeloperoxidase confers specificity to vascular ECM proteins as targets of tyrosine nitration. J Clin Invest. 2001; 108: 1759–1770.CrossrefMedlineGoogle Scholar30 Brennan ML, Wu W, Fu X, Shen Z, Song W, Frost H, Vadseth C, Narine L, Lenkiewicz E, Borchers MT, Lusis AJ, Lee JJ, Lee NA, Abu-Soud HM, Ischiropoulos H, Hazen SL. A tale of two controversies: defining both the role of peroxidases in nitrotyrosine formation in vivo using eosinophil peroxidase and myeloperoxidase-deficient mice, and the nature of peroxidase-generated reactive nitrogen species. J Biol Chem. 2002; 277: 17415–17427.CrossrefMedlineGoogle Scholar31 Gaut JP, Byun J, Tran HD, Lauber WM, Carroll JA, Hotchkiss RS, Belaaouaj A, Heinecke JW. Myeloperoxidase produces nitrating oxidants in vivo. J Clin Invest. 2002; 109: 1311–1319.CrossrefMedlineGoogle Scholar32 Vita JA, Brennan ML, Gokce N, Mann SA, Goormastic M, Shishehbor MH, Penn MS, Keaney JF, Hazen SL. Serum myeloperoxidase level independently predicts endothelial dysfunction in humans. Circulation. 2004. (In press.)Google Scholar33 Kutter D, Devaquet P, Vanderstocken G, et al. Consequences of total and subtotal myeloperoxidase deficiency: risk or benefit? Acta Haematol. 2000; 104: 10–15.CrossrefMedlineGoogle Scholar34 Nikpoor B, Turecki G, Fournier C, Theroux P, Rouleau GA. A functional myeloperoxidase polymorphic variant is associated with coronary artery disease in French-Canadians. Am Heart J. 2001; 142: 336–339.CrossrefMedlineGoogle Scholar35 Pecoits-Filho R, Stenvinkel P, Marchlewska A, Heimburger O, Barany P, Hoff CM, Holmes CJ, Suliman M, Lindholm B, Schalling M, Nordfors L. A functional variant of the myeloperoxidase gene is associated with cardiovascular disease in end-stage renal disease patients. Kidney Int Suppl. 2003; 84: 172–176.Google Scholar36 Asselbergs FW, Tervaert JW, Tio RA. Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med. 2004; 350: 516–518.CrossrefGoogle Scholar37 Fu X, Kassim SY, Parks WC, Heinecke JW. Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (matrix metalloproteinase (MMP)-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem. 2001; 276: 41279–41287.CrossrefMedlineGoogle Scholar38 Kettle AJ, Winterbourn CC. Assays for the chlorination activity of myeloperoxidase. Methods Enzymol. 1994; 233: 502–512.CrossrefMedlineGoogle Scholar39 Napoli C, Ignarro LJ. Nitric oxide and atherosclerosis. Nitric Oxide. 2001; 5: 88–97.CrossrefMedlineGoogle Scholar40 Vita JA, Keaney JF, Jr. Endothelial function: a barometer for cardiovascular risk? Circulation. 2002; 106: 640–642.LinkGoogle Scholar41 van der Wal AC, Becker AE, van der Loos CM, Das PK. Site of intimal rupture or erosion of thrombosed coronary atherosclerotic plaques is characterized by an inflammatory process irrespective of the dominant plaque morphology. Circulation. 1994; 89: 36–44.CrossrefMedlineGoogle Scholar42 Kolodgie FD, Burke AP, Farb A, Weber DK, Kutys R, Wight TN, Virmani R. Differential accumulation of proteoglycans and hyaluronan in culprit lesions: insights into plaque erosion. Arterioscler Thromb Vasc Biol. 2002; 22: 1642–1648.LinkGoogle Scholar43 Tricot O, Mallat Z, Heymes C, Belmin J, Leseche G, Tedgui A. Relation between endothelial cell apoptosis and blood flow direction in human atherosclerotic plaques. Circulation. 2000; 101: 2450–2453.CrossrefMedlineGoogle Scholar44 Mutin M, Canavy I, Blam A, Bory M, Sampol J, Dignat-George F. Direct evidence of endothelial injury in acute myocardial infarction and unstable angina by demonstration of circulating endothelial cells. Blood. 1999; 93: 2951–2958.CrossrefMedlineGoogle Scholar45 Penn MS, Patel CV, Cui MZ, DiCorleto PE, Chisolm GM. LDL increases inactive tissue factor on vascular smooth muscle cell surfaces: hydrogen peroxide activates latent cell surface tissue factor. Circulation. 1999; 99: 1753–1759.CrossrefMedlineGoogle Scholar46 Penn MS, Cui MZ, Winokur AL, Bethea J, Hamilton TA, DiCorleto PE, Chisolm GM. Smooth muscle cell surface tissue factor pathway activation by oxidized low-density lipoprotein requires cellular lipid peroxidation. Blood. 2000; 96: 3056–3063.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Adlam D, Zarebinski M, Uren N, Ptaszynski P, Oldroyd K, Munir S, Zaman A, Contractor H, Kiss R, Édes I, Szachniewicz J, Nagy G, Garcia M, Tomcsanyi J, Irving J, Sharp A, Musialek P, Lupkovics G, Shirodaria C, Selvanayagam J, Quinn P, Ng L, Roth M, Insko M, Haber B, Hill S, Siegel L, Tulloch S and Channon K (2022) A Randomized, double-blind, dose ranging clinical trial of intravenous FDY-5301 in acute STEMI patients undergoing primary PCI, International Journal of Cardiology, 10.1016/j.ijcard.2021.11.016, 347, (1-7), Online publication date: 1-Jan-2022. Roumeliotis S, Veljkovic A, Georgianos P, Lazarevic G, Perisic Z, Hadzi-Djokic J, Liakopoulos V, Kocic G and Ciobica A (2021) Association between Biomarkers of Oxidative Stress and Inflammation with Cardiac Necrosis and Heart Failure in Non-ST Segment Elevation Myocardial Infarction Patients and Various Degrees of Kidney Function, Oxidative Medicine and Cellular Longevity, 10.1155/2021/3090120, 2021, (1-12), Online publication date: 1-Nov-2021. Lee A, Wang Y, Chang S, Lo P, Chang C, Lu J, Burns A, Chen C, Kakino A, Sawamura T and Chang K (2020) Detection of a High Ratio of Soluble to Membrane‐Bound LOX‐1 in Aspirated Coronary Thrombi From Patients With ST‐Segment–Elevation Myocardial Infarction, Journal of the American Heart Association, 9:2, Online publication date: 21-Jan-2020. Simsek B, Yanar K and Çakatay U Proatherogenic Importance of Carbamylation-induced Protein Damage and Type 2 Diabetes Mellitus: A Systematic Review, Current Diabetes Reviews, 10.2174/1573399816666200107102918, 16:6, (608-618) Gamon L, Dieterich S, Ignasiak M, Schrameyer V and Davies M (2020) Iodide modulates protein damage induced by the inflammation-associated heme enzyme myeloperoxidase, Redox Biology, 10.1016/j.redox.2019.101331, 28, (101331), Online publication date: 1-Jan-2020. Yamamoto H, Yoshida N, Shinke T, Otake H, Kuroda M, Sakaguchi K, Hirota Y, Toba T, Takahashi H, Terashita D, Uzu K, Tahara N, Shinkura Y, Kuroda K, Nagasawa Y, Nagano Y, Tsukiyama Y, Yanaka K, Emoto T, Sasaki N, Yamashita T, Ogawa W and Hirata K (2018) Impact of CD14 ++ CD16 + monocytes on coronary plaque vulnerability assessed by optical coherence tomography in coronary artery disease patients, Atherosclerosis, 10.1016/j.atherosclerosis.2018.01.010, 269, (245-251), Online publication date: 1-Feb-2018. Desikan R, Narasimhulu C, Khan B, Rajagopalan S and Parthasarathy S (2017) Myeloperoxidase (MPO): Do We Need Inhibitors? Mechanisms of Vascular Defects in Diabetes Mellitus, 10.1007/978-3-319-60324-7_24, (535-571), . Soubhye J, Chikh Alard I, Aldib I, Prévost M, Gelbcke M, De Carvalho A, Furtmüller P, Obinger C, Flemmig J, Tadrent S, Meyer F, Rousseau A, Nève J, Mathieu V, Zouaoui Boudjeltia K, Dufrasne F and Van Antwerpen P (2017) Discovery of Novel Potent Reversible and Irreversible Myeloperoxidase Inhibitors Using Virtual Screening Procedure, Journal of Medicinal Chemistry, 10.1021/acs.jmedchem.7b00285, 60:15, (6563-6586), Online publication date: 10-Aug-2017. Li Y, Wang H, Qian J, Kim H, Wu J, Wang L, Zhou C, Yang Z and Lu X (2017) PRISMA-combined Myeloperoxidase -463G/A gene polymorphism and coronary artery disease, Medicine, 10.1097/MD.0000000000006461, 96:12, (e6461), Online publication date: 1-Mar-2017. Yoshida N, Yamamoto H, Shinke T, Otake H, Kuroda M, Terashita D, Takahashi H, Sakaguchi K, Hirota Y, Emoto T, Amin H, Mizoguchi T, Hayashi T, Sasaki N, Yamashita T, Ogawa W and Hirata K (2017) Impact of CD14++CD16+ monocytes on plaque vulnerability in diabetic and non-diabetic patients with asymptomatic coronary artery disease: a cross-sectional study, Cardiovascular Diabetology, 10.1186/s12933-017-0577-8, 16:1, Online publication date: 1-Dec-2017. Hayat U, Thondapu V, Tsay T and Barlis P (2016) Atherogenesis and Inflammation Interventional Cardiology, 10.1002/9781118983652.ch1, (1-16) Jaw J, Tsuruta M, Oh Y, Schipilow J, Hirano Y, Ngan D, Suda K, Li Y, Oh J, Moritani K, Tam S, Ford N, van Eeden S, Wright J, Man S and Sin D (2016) Lung exposure to lipopolysaccharide causes atherosclerotic plaque destabilisation, European Respiratory Journal, 10.1183/13993003.00972-2015, 48:1, (205-215), Online publication date: 1-Jul-2016. Tiyerili V, Camara B, Becher M, Schrickel J, Lütjohann D, Mollenhauer M, Baldus S, Nickenig G and Andrié R (2016) Neutrophil-derived myeloperoxidase promotes atherogenesis and neointima formation in mice, International Journal of Cardiology, 10.1016/j.ijcard.2015.11.128, 204, (29-36), Online publication date: 1-Feb-2016. Gu X, Huang Y, Levison B, Gerstenecker G, DiDonato A, Hazen L, Lee J, Gogonea V, DiDonato J and Hazen S (2016) Identification of Critical Paraoxonase 1 Residues Involved in High Density Lipoprotein Interaction, Journal of Biological Chemistry, 10.1074/jbc.M115.678334, 291:4, (1890-1904), Online publication date: 1-Jan-2016. Soubhye J, Meyer F, Furtmüller P, Obinger C, Dufrasne F and Antwerpen P (2016) Characterization of chemical features of potent myeloperoxidase inhibitors, Future Medicinal Chemistry, 10.4155/fmc-2016-0031, 8:11, (1163-1177), Online publication date: 1-Jul-2016. Okamura D and Pennathur S (2015) The balance of powers: Redox regulation of fibrogenic pathways in kidney injury, Redox Biology, 10.1016/j.redox.2015.09.039, 6, (495-504), Online publication date: 1-Dec-2015. Koch C, Henrich M and Heidt M (2014) Sequential Analysis of Myeloperoxidase for Prediction of Adverse Events After Suspected Acute Coronary Ischemia, Clinical Cardiology, 10.1002/clc.22336, 37:12, (744-749), Online publication date: 1-Dec-2014. Pop-Busui R, Mehta M and Pennathur S (2014) Oxidative Stress and Cardiovascular Disease in Diabetes Studies in Diabetes, 10.1007/978-1-4899-8035-9_11, (189-235), . Koeth R, Kalantar-Zadeh K, Wang Z, Fu X, Tang W and Hazen S (2013) Protein Carbamylation Predicts Mortality in ESRD, Journal of the American Society of Nephrology, 10.1681/ASN.2012030254, 24:5, (853-861), Online publication date: 1-May-2013. Hou Z, Lu B, Gao Y, Cao H, Yu F, Jing N, Chen X, Cong X, Roy S and Budoff M (2013) Matrix Metalloproteinase-9 (MMP-9) and Myelo

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call