Skip to main content
Top
Published in: Current Cardiovascular Imaging Reports 1/2011

01-02-2011

Imaging Myeloperoxidase Activity in Cardiovascular Disease

Author: John A. Ronald

Published in: Current Cardiovascular Imaging Reports | Issue 1/2011

Login to get access

Abstract

Inflammation and oxidative stress play central roles in the pathogenesis of atherosclerosis and outcomes such as myocardial infarction and stroke. The enzyme myeloperoxidase (MPO) is secreted by activated leukocytes in atherosclerotic plaques and generates several pro-oxidative species that participate in biological activities that promote plaque destabilization and rupture; the catastrophic event that defines which plaques are benign and which are life-threatening. Serum MPO levels are also highly predictive of downstream cardiovascular events and patient outcome. Recently, several molecular imaging probes sensitive to the presence of MPO activity have been developed. Here we review the design and application of probes developed for MRI, single photon emission CT, and fluorescence and bioluminescence imaging. Continued development of these probes may one day allow detection of plaques prior to plaque rupture, yielding significant diagnostic and prognostic improvements and ultimately allowing proper staging and treatment of high-risk patients.
Literature
1.
go back to reference Hackam DG, Anand SS: Emerging risk factors for atherosclerotic vascular disease: a critical review of the evidence. JAMA 2003, 290(7):932–940.CrossRefPubMed Hackam DG, Anand SS: Emerging risk factors for atherosclerotic vascular disease: a critical review of the evidence. JAMA 2003, 290(7):932–940.CrossRefPubMed
3.
go back to reference Libby P, Aikawa M: Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med 2002, 8(11):1257–1262.CrossRefPubMed Libby P, Aikawa M: Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med 2002, 8(11):1257–1262.CrossRefPubMed
4.
go back to reference Casscells W, Naghavi M, Willerson JT: Vulnerable atherosclerotic plaque: a multifocal disease. Circulation 2003, 107(16):2072–2075.CrossRefPubMed Casscells W, Naghavi M, Willerson JT: Vulnerable atherosclerotic plaque: a multifocal disease. Circulation 2003, 107(16):2072–2075.CrossRefPubMed
5.
go back to reference Falk E: Why do plaques rupture? Circulation 1992, 86(6 Suppl):III30–42.PubMed Falk E: Why do plaques rupture? Circulation 1992, 86(6 Suppl):III30–42.PubMed
6.
go back to reference Naghavi M, Libby P, Falk E, et al.: From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part II. Circulation 2003, 108(15):1772–1778.CrossRefPubMed Naghavi M, Libby P, Falk E, et al.: From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part II. Circulation 2003, 108(15):1772–1778.CrossRefPubMed
7.
go back to reference Naghavi M, Libby P, Falk E, et al.: From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part I. Circulation 2003, 108(14):1664–1672.CrossRefPubMed Naghavi M, Libby P, Falk E, et al.: From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part I. Circulation 2003, 108(14):1664–1672.CrossRefPubMed
8.
go back to reference Fuster V, Moreno PR, Fayad ZA, et al.: Atherothrombosis and high-risk plaque: part I: evolving concepts. J Am Coll Cardiol 2005, 46(6):937–954.CrossRefPubMed Fuster V, Moreno PR, Fayad ZA, et al.: Atherothrombosis and high-risk plaque: part I: evolving concepts. J Am Coll Cardiol 2005, 46(6):937–954.CrossRefPubMed
9.
go back to reference Geng Y-J, Libby P: Progression of atheroma: a struggle between death and procreation. Arteriosclerosis, Thrombosis, and Vascular Biology 2002, 22(9):1370–1380.CrossRefPubMed Geng Y-J, Libby P: Progression of atheroma: a struggle between death and procreation. Arteriosclerosis, Thrombosis, and Vascular Biology 2002, 22(9):1370–1380.CrossRefPubMed
10.
go back to reference Moreno PR, Purushothaman KR, Fuster V, et al.: Plaque neovascularization is increased in ruptured atherosclerotic lesions of human aorta: implications for plaque vulnerability. Circulation 2004, 110(14):2032–2038.CrossRefPubMed Moreno PR, Purushothaman KR, Fuster V, et al.: Plaque neovascularization is increased in ruptured atherosclerotic lesions of human aorta: implications for plaque vulnerability. Circulation 2004, 110(14):2032–2038.CrossRefPubMed
11.
go back to reference Galis ZS, Sukhova GK, Lark MW, Libby P: Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest 1994, 94(6):2493–2503.CrossRefPubMed Galis ZS, Sukhova GK, Lark MW, Libby P: Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest 1994, 94(6):2493–2503.CrossRefPubMed
12.
go back to reference Nicholls SJ, Hazen SL: Myeloperoxidase and cardiovascular disease. Arteriosclerosis, Thrombosis, and Vascular Biology 2005, 25(6):1102–1111.CrossRefPubMed Nicholls SJ, Hazen SL: Myeloperoxidase and cardiovascular disease. Arteriosclerosis, Thrombosis, and Vascular Biology 2005, 25(6):1102–1111.CrossRefPubMed
14.
go back to reference Schultz J, Kaminker K: Myeloperoxidase of the leucocyte of normal human blood. I. Content and localization. Arch Biochem Biophys 1962, 96:465–467.CrossRef Schultz J, Kaminker K: Myeloperoxidase of the leucocyte of normal human blood. I. Content and localization. Arch Biochem Biophys 1962, 96:465–467.CrossRef
15.
go back to reference Bos A, Wever R, Roos D: Characterization and quantification of the peroxidase in human monocytes. Biochim Biophys Acta 1978, 525(1):37–44.PubMed Bos A, Wever R, Roos D: Characterization and quantification of the peroxidase in human monocytes. Biochim Biophys Acta 1978, 525(1):37–44.PubMed
16.
go back to reference 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(1):437–444.CrossRefPubMed 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(1):437–444.CrossRefPubMed
17.
go back to reference Sugiyama S, Okada Y, Sukhova GK, et al.: Macrophage myeloperoxidase regulation by granulocyte macrophage colony-stimulating factor in human atherosclerosis and implications in acute coronary syndromes. Am J Pathol 2001, 158(3):879–891.PubMed Sugiyama S, Okada Y, Sukhova GK, et al.: Macrophage myeloperoxidase regulation by granulocyte macrophage colony-stimulating factor in human atherosclerosis and implications in acute coronary syndromes. Am J Pathol 2001, 158(3):879–891.PubMed
18.
go back to reference Hazen SL, Zhang R, Shen Z, et al.: 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(10):950–958.PubMed Hazen SL, Zhang R, Shen Z, et al.: 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(10):950–958.PubMed
19.
go back to reference Nicholls SJ, Zheng L, Hazen SL: Formation of dysfunctional high-density lipoprotein by myeloperoxidase. Trends Cardiovasc Med 2005, 15(6):212–219.CrossRefPubMed Nicholls SJ, Zheng L, Hazen SL: Formation of dysfunctional high-density lipoprotein by myeloperoxidase. Trends Cardiovasc Med 2005, 15(6):212–219.CrossRefPubMed
20.
go back to reference Nicholls SJ, Hazen SL: Myeloperoxidase, modified lipoproteins, and atherogenesis. J Lipid Res 2009, 50 Suppl:S346–351.CrossRefPubMed Nicholls SJ, Hazen SL: Myeloperoxidase, modified lipoproteins, and atherogenesis. J Lipid Res 2009, 50 Suppl:S346–351.CrossRefPubMed
21.
go back to reference • Tavora FR, Ripple M, Li L, Burke AP: Monocytes and neutrophils expressing myeloperoxidase occur in fibrous caps and thrombi in unstable coronary plaques. BMC Cardiovasc Disord 2009, 9:27. This article clearly shows that MPO is expressed in plaques with histological characteristics of vulnerable plaques (thin-cap atheroma), and absent from more stable plaques (fibroatheroma). It also demonstrates that thrombi associated with disrupted plaques contain appreciable numbers of MPO-positive cells.CrossRefPubMed • Tavora FR, Ripple M, Li L, Burke AP: Monocytes and neutrophils expressing myeloperoxidase occur in fibrous caps and thrombi in unstable coronary plaques. BMC Cardiovasc Disord 2009, 9:27. This article clearly shows that MPO is expressed in plaques with histological characteristics of vulnerable plaques (thin-cap atheroma), and absent from more stable plaques (fibroatheroma). It also demonstrates that thrombi associated with disrupted plaques contain appreciable numbers of MPO-positive cells.CrossRefPubMed
22.
go back to reference Marsche G, Hammer A, Oskolkova O, et al.: Hypochlorite-modified high density lipoprotein, a high affinity ligand to scavenger receptor class B, type I, impairs high density lipoprotein-dependent selective lipid uptake and reverse cholesterol transport. J Biol Chem 2002, 277(35):32172–32179.CrossRefPubMed Marsche G, Hammer A, Oskolkova O, et al.: Hypochlorite-modified high density lipoprotein, a high affinity ligand to scavenger receptor class B, type I, impairs high density lipoprotein-dependent selective lipid uptake and reverse cholesterol transport. J Biol Chem 2002, 277(35):32172–32179.CrossRefPubMed
23.
go back to reference Hazell LJ, Arnold L, Flowers D, et al.: Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest 1996, 97(6):1535–1544.CrossRefPubMed Hazell LJ, Arnold L, Flowers D, et al.: Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest 1996, 97(6):1535–1544.CrossRefPubMed
24.
go back to reference 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(9):2075–2081.CrossRefPubMed 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(9):2075–2081.CrossRefPubMed
25.
go back to reference Wang Z, Nicholls SJ, Rodriguez ER, et al.: Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 2007, 13(10):1176–1184.CrossRefPubMed Wang Z, Nicholls SJ, Rodriguez ER, et al.: Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 2007, 13(10):1176–1184.CrossRefPubMed
26.
go back to reference Eiserich JP, Baldus S, Brennan M-L, et al.: Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Science 2002, 296(5577):2391–2394.CrossRefPubMed Eiserich JP, Baldus S, Brennan M-L, et al.: Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Science 2002, 296(5577):2391–2394.CrossRefPubMed
27.
go back to reference Sugiyama S, Kugiyama K, Aikawa M, et al.: Hypochlorous acid, a macrophage product, induces endothelial apoptosis and tissue factor expression: involvement of myeloperoxidase-mediated oxidant in plaque erosion and thrombogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 2004, 24(7):1309–1314.CrossRefPubMed Sugiyama S, Kugiyama K, Aikawa M, et al.: Hypochlorous acid, a macrophage product, induces endothelial apoptosis and tissue factor expression: involvement of myeloperoxidase-mediated oxidant in plaque erosion and thrombogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 2004, 24(7):1309–1314.CrossRefPubMed
28.
go back to reference Fu X, Kassim SY, Parks WC, Heinecke JW: Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem 2001, 276(44):41279–41287.CrossRefPubMed Fu X, Kassim SY, Parks WC, Heinecke JW: Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem 2001, 276(44):41279–41287.CrossRefPubMed
29.
go back to reference Zhang R, Brennan ML, Fu X, et al.: Association between myeloperoxidase levels and risk of coronary artery disease. JAMA 2001, 286(17):2136–2142.CrossRefPubMed Zhang R, Brennan ML, Fu X, et al.: Association between myeloperoxidase levels and risk of coronary artery disease. JAMA 2001, 286(17):2136–2142.CrossRefPubMed
30.
go back to reference Brennan M-L, Penn MS, Van Lente F, et al.: Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med 2003, 349(17):1595–1604.CrossRefPubMed Brennan M-L, Penn MS, Van Lente F, et al.: Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med 2003, 349(17):1595–1604.CrossRefPubMed
31.
go back to reference • Meuwese MC, Stroes ESG, Hazen SL, et al.: Serum myeloperoxidase levels are associated with the future risk of coronary artery disease in apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. J Am Coll Cardiol 2007, 50(2):159–165. This article shows that elevated MPO serum levels are predictive of future risk of CAD during an 8-year follow-up in apparently healthy individuals. This suggests that inflammatory activation, as detected by MPO secretion, precedes CAD onset by many years.CrossRefPubMed • Meuwese MC, Stroes ESG, Hazen SL, et al.: Serum myeloperoxidase levels are associated with the future risk of coronary artery disease in apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. J Am Coll Cardiol 2007, 50(2):159–165. This article shows that elevated MPO serum levels are predictive of future risk of CAD during an 8-year follow-up in apparently healthy individuals. This suggests that inflammatory activation, as detected by MPO secretion, precedes CAD onset by many years.CrossRefPubMed
32.
go back to reference Wong ND, Gransar H, Narula J, et al.: Myeloperoxidase, subclinical atherosclerosis, and cardiovascular disease events. JACC Cardiovasc Imaging 2009, 2(9):1093–1099.CrossRefPubMed Wong ND, Gransar H, Narula J, et al.: Myeloperoxidase, subclinical atherosclerosis, and cardiovascular disease events. JACC Cardiovasc Imaging 2009, 2(9):1093–1099.CrossRefPubMed
33.
go back to reference Touzé E, Toussaint J-F, Coste J, et al.: Reproducibility of high-resolution MRI for the identification and the quantification of carotid atherosclerotic plaque components: consequences for prognosis studies and therapeutic trials. Stroke 2007, 38(6):1812–1819.CrossRefPubMed Touzé E, Toussaint J-F, Coste J, et al.: Reproducibility of high-resolution MRI for the identification and the quantification of carotid atherosclerotic plaque components: consequences for prognosis studies and therapeutic trials. Stroke 2007, 38(6):1812–1819.CrossRefPubMed
34.
go back to reference Bogdanov A, Matuszewski L, Bremer C, et al.: Oligomerization of paramagnetic substrates result in signal amplification and can be used for MR imaging of molecular targets. Mol Imaging 2002, 1(1):16–23.CrossRefPubMed Bogdanov A, Matuszewski L, Bremer C, et al.: Oligomerization of paramagnetic substrates result in signal amplification and can be used for MR imaging of molecular targets. Mol Imaging 2002, 1(1):16–23.CrossRefPubMed
35.
go back to reference Chen JW, Pham W, Weissleder R, Bogdanov A: Human myeloperoxidase: a potential target for molecular MR imaging in atherosclerosis. Magn Reson Med 2004, 52(5):1021–1028.CrossRefPubMed Chen JW, Pham W, Weissleder R, Bogdanov A: Human myeloperoxidase: a potential target for molecular MR imaging in atherosclerosis. Magn Reson Med 2004, 52(5):1021–1028.CrossRefPubMed
36.
go back to reference Dunford HB, Hsuanyu Y: Kinetics of oxidation of serotonin by myeloperoxidase compounds I and II. Biochem Cell Biol 1999, 77(5):449–457.CrossRefPubMed Dunford HB, Hsuanyu Y: Kinetics of oxidation of serotonin by myeloperoxidase compounds I and II. Biochem Cell Biol 1999, 77(5):449–457.CrossRefPubMed
37.
go back to reference Querol M, Chen JW, Bogdanov AA: A paramagnetic contrast agent with myeloperoxidase-sensing properties. Org Biomol Chem 2006, 4(10):1887–1895.CrossRefPubMed Querol M, Chen JW, Bogdanov AA: A paramagnetic contrast agent with myeloperoxidase-sensing properties. Org Biomol Chem 2006, 4(10):1887–1895.CrossRefPubMed
38.
go back to reference Querol M, Chen JW, Weissleder R, Bogdanov A: DTPA-bisamide-based MR sensor agents for peroxidase imaging. Org Lett 2005, 7(9):1719–1722.CrossRefPubMed Querol M, Chen JW, Weissleder R, Bogdanov A: DTPA-bisamide-based MR sensor agents for peroxidase imaging. Org Lett 2005, 7(9):1719–1722.CrossRefPubMed
39.
go back to reference • Rodríguez E, Nilges M, Weissleder R, Chen JW: Activatable magnetic resonance imaging agents for myeloperoxidase sensing: mechanism of activation, stability, and toxicity. J Am Chem Soc 2010, 132(1):168–177. This study details the mechanisms of activation, specificity, stability, and cytotoxicity characteristics of several MRI MPO probes, including MPO-Gd.CrossRefPubMed • Rodríguez E, Nilges M, Weissleder R, Chen JW: Activatable magnetic resonance imaging agents for myeloperoxidase sensing: mechanism of activation, stability, and toxicity. J Am Chem Soc 2010, 132(1):168–177. This study details the mechanisms of activation, specificity, stability, and cytotoxicity characteristics of several MRI MPO probes, including MPO-Gd.CrossRefPubMed
40.
go back to reference Chen JW, Querol Sans M, Bogdanov A, Weissleder R: Imaging of myeloperoxidase in mice by using novel amplifiable paramagnetic substrates. Radiology 2006, 240(2):473–481.CrossRefPubMed Chen JW, Querol Sans M, Bogdanov A, Weissleder R: Imaging of myeloperoxidase in mice by using novel amplifiable paramagnetic substrates. Radiology 2006, 240(2):473–481.CrossRefPubMed
41.
go back to reference •• Nahrendorf M, Sosnovik D, Chen JW, et al.: Activatable magnetic resonance imaging agent reports myeloperoxidase activity in healing infarcts and noninvasively detects the antiinflammatory effects of atorvastatin on ischemia-reperfusion injury. Circulation 2008, 117(9):1153–1160. This is the first study showing the ability to image MPO activity using the MRI probe MPO-Gd in a mouse model of cardiovascular disease (myocardial ischemia-reperfusion injury). Furthermore, the anti-inflammatory effects of atorvastatin treatment were successfully tracked.CrossRefPubMed •• Nahrendorf M, Sosnovik D, Chen JW, et al.: Activatable magnetic resonance imaging agent reports myeloperoxidase activity in healing infarcts and noninvasively detects the antiinflammatory effects of atorvastatin on ischemia-reperfusion injury. Circulation 2008, 117(9):1153–1160. This is the first study showing the ability to image MPO activity using the MRI probe MPO-Gd in a mouse model of cardiovascular disease (myocardial ischemia-reperfusion injury). Furthermore, the anti-inflammatory effects of atorvastatin treatment were successfully tracked.CrossRefPubMed
42.
go back to reference Chen JW, Breckwoldt MO, Aikawa E, Chiang G, Weissleder R: Myeloperoxidase-targeted imaging of active inflammatory lesions in murine experimental autoimmune encephalomyelitis. Brain 2008, 131(Pt 4):1123–1133.CrossRefPubMed Chen JW, Breckwoldt MO, Aikawa E, Chiang G, Weissleder R: Myeloperoxidase-targeted imaging of active inflammatory lesions in murine experimental autoimmune encephalomyelitis. Brain 2008, 131(Pt 4):1123–1133.CrossRefPubMed
43.
go back to reference •• Breckwoldt MO, Chen JW, Stangenberg L, et al.: Tracking the inflammatory response in stroke in vivo by sensing the enzyme myeloperoxidase. Proc Natl Acad Sci USA 2008, 105(47):18584–18589. This study showed that MPO-Gd can be used to track the temporal changes in inflammatory response and oxidative stress in a mouse model of stroke.CrossRefPubMed •• Breckwoldt MO, Chen JW, Stangenberg L, et al.: Tracking the inflammatory response in stroke in vivo by sensing the enzyme myeloperoxidase. Proc Natl Acad Sci USA 2008, 105(47):18584–18589. This study showed that MPO-Gd can be used to track the temporal changes in inflammatory response and oxidative stress in a mouse model of stroke.CrossRefPubMed
44.
go back to reference •• Ronald JA, Chen JW, Chen Y, et al.: Enzyme-sensitive magnetic resonance imaging targeting myeloperoxidase identifies active inflammation in experimental rabbit atherosclerotic plaques. Circulation 2009, 120(7):592–599. This is currently the only study showing the ability to noninvasively detect MPO activity in an animal model of atherosclerosis in vivo. It required the combined use of a rabbit model of atherosclerosis, high-resolution MRI, and the activatable MRI probe, MPO-Gd.CrossRefPubMed •• Ronald JA, Chen JW, Chen Y, et al.: Enzyme-sensitive magnetic resonance imaging targeting myeloperoxidase identifies active inflammation in experimental rabbit atherosclerotic plaques. Circulation 2009, 120(7):592–599. This is currently the only study showing the ability to noninvasively detect MPO activity in an animal model of atherosclerosis in vivo. It required the combined use of a rabbit model of atherosclerosis, high-resolution MRI, and the activatable MRI probe, MPO-Gd.CrossRefPubMed
45.
go back to reference Brennan ML, Anderson MM, Shih DM, et al.: Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest 2001, 107(4):419–430.CrossRefPubMed Brennan ML, Anderson MM, Shih DM, et al.: Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest 2001, 107(4):419–430.CrossRefPubMed
46.
go back to reference Querol Sans M, Chen JW, Weissleder R, Bogdanov AA: Myeloperoxidase activity imaging using (67)Ga labeled substrate. Mol Imaging Biol 2005, 7(6):403–410.CrossRefPubMed Querol Sans M, Chen JW, Weissleder R, Bogdanov AA: Myeloperoxidase activity imaging using (67)Ga labeled substrate. Mol Imaging Biol 2005, 7(6):403–410.CrossRefPubMed
47.
go back to reference Li D, Patel AR, Klibanov AL, et al.: Molecular imaging of atherosclerotic plaques targeted to oxidized LDL receptor LOX-1 by SPECT/CT and magnetic resonance. Circ Cardiovasc Imaging 2010, 3(4):464–472.CrossRefPubMed Li D, Patel AR, Klibanov AL, et al.: Molecular imaging of atherosclerotic plaques targeted to oxidized LDL receptor LOX-1 by SPECT/CT and magnetic resonance. Circ Cardiovasc Imaging 2010, 3(4):464–472.CrossRefPubMed
48.
go back to reference Silvera SS, Aidi HE, Rudd JHF, et al.: Multimodality imaging of atherosclerotic plaque activity and composition using FDG-PET/CT and MRI in carotid and femoral arteries. Atherosclerosis 2009, 207(1):139–143.CrossRefPubMed Silvera SS, Aidi HE, Rudd JHF, et al.: Multimodality imaging of atherosclerotic plaque activity and composition using FDG-PET/CT and MRI in carotid and femoral arteries. Atherosclerosis 2009, 207(1):139–143.CrossRefPubMed
49.
go back to reference •• Shepherd J, Hilderbrand SA, Waterman P, et al.: A fluorescent probe for the detection of myeloperoxidase activity in atherosclerosis-associated macrophages. Chem Biol 2007, 14(11):1221–1231. This study describes the first in vivo imaging of MPO activity using an activatable fluorescent probe called SNAPF. Upon activation the agent emits far-red light, making it highly suitable for in vivo imaging.CrossRefPubMed •• Shepherd J, Hilderbrand SA, Waterman P, et al.: A fluorescent probe for the detection of myeloperoxidase activity in atherosclerosis-associated macrophages. Chem Biol 2007, 14(11):1221–1231. This study describes the first in vivo imaging of MPO activity using an activatable fluorescent probe called SNAPF. Upon activation the agent emits far-red light, making it highly suitable for in vivo imaging.CrossRefPubMed
50.
go back to reference Calfon MA, Vinegoni C, Ntziachristos V, Jaffer FA: Intravascular near-infrared fluorescence molecular imaging of atherosclerosis: toward coronary arterial visualization of biologically high-risk plaques. J Biomed Opt 2010, 15(1):011107.CrossRefPubMed Calfon MA, Vinegoni C, Ntziachristos V, Jaffer FA: Intravascular near-infrared fluorescence molecular imaging of atherosclerosis: toward coronary arterial visualization of biologically high-risk plaques. J Biomed Opt 2010, 15(1):011107.CrossRefPubMed
51.
go back to reference •• Gross S, Gammon ST, Moss BL, et al.: Bioluminescence imaging of myeloperoxidase activity in vivo. Nat Med 2009, 15(4):455–461. This study describes the use of the chemiluminescent agent luminol to locate MPO activity in vivo in a variety of mouse models using bioluminescence imaging. In vivo specificity of luminol oxidation (and thus luminescence) to MPO, but not eosinophil peroxidase, activity is also shown.CrossRefPubMed •• Gross S, Gammon ST, Moss BL, et al.: Bioluminescence imaging of myeloperoxidase activity in vivo. Nat Med 2009, 15(4):455–461. This study describes the use of the chemiluminescent agent luminol to locate MPO activity in vivo in a variety of mouse models using bioluminescence imaging. In vivo specificity of luminol oxidation (and thus luminescence) to MPO, but not eosinophil peroxidase, activity is also shown.CrossRefPubMed
Metadata
Title
Imaging Myeloperoxidase Activity in Cardiovascular Disease
Author
John A. Ronald
Publication date
01-02-2011
Publisher
Current Science Inc.
Published in
Current Cardiovascular Imaging Reports / Issue 1/2011
Print ISSN: 1941-9066
Electronic ISSN: 1941-9074
DOI
https://doi.org/10.1007/s12410-010-9056-2

Other articles of this Issue 1/2011

Current Cardiovascular Imaging Reports 1/2011 Go to the issue