251
|
Csató V, Pető A, Fülöp GÁ, Rutkai I, Pásztor ET, Fagyas M, Kalász J, Édes I, Tóth A, Papp Z. Myeloperoxidase evokes substantial vasomotor responses in isolated skeletal muscle arterioles of the rat. Acta Physiol (Oxf) 2015; 214:109-23. [PMID: 25760778 PMCID: PMC4654238 DOI: 10.1111/apha.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/19/2015] [Accepted: 03/09/2015] [Indexed: 12/20/2022]
Abstract
Aims Myeloperoxidase (MPO) catalyses the formation of a wide variety of oxidants, including hypochlorous acid (HOCl), and contributes to cardiovascular disease progression. We hypothesized that during its action MPO evokes substantial vasomotor responses. Methods Following exposure to MPO (1.92 mU mL−1) in the presence of increasing concentrations of hydrogen peroxide (H2O2), changes in arteriolar diameter of isolated gracilis skeletal muscle arterioles (SMAs) and coronary arterioles (CAs) and in the isometric force in basilar arteries (BAs) of the rat were monitored. Results Myeloperoxidase increased vascular tone to different degrees in CAs, SMAs and BAs. The mechanism of increased vasoconstriction was studied in detail in SMAs. MPO-evoked vasoconstrictions were prevented by the MPO inhibitor 4-aminobenzhydrazide (50 μm), by endothelium removal in the SMAs. Surprisingly, the HOCl scavenger L-methionine (100 μm), the thromboxane A2 (TXA2) antagonist SQ-29548 (1 μm) or the non-specific cyclooxygenase (COX) antagonist indomethacin (1 μm) converted the MPO-evoked vasoconstrictions to pronounced vasodilations in SMAs, not seen in the presence of H2O2. In contrast to noradrenaline-induced vasoconstrictions, the MPO-evoked vasoconstrictions were not accompanied by significant increases in arteriolar [Ca2+] levels in SMAs. Conclusion These data showed that H2O2-derived HOCl to be a potent vasoconstrictor upon MPO application. HOCl activated the COX pathway, causing the synthesis and release of a TXA2-like substance to increase the Ca2+ sensitivity of the contractile apparatus in vascular smooth muscle cells and thereby to augment H2O2-evoked vasoconstrictions. Nevertheless, inhibition of the HOCl–COX–TXA2 pathway unmasked the effects of additional MPO-derived radicals with a marked vasodilatory potential in SMAs.
Collapse
Affiliation(s)
- V. Csató
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - A. Pető
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - G. Á. Fülöp
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - I. Rutkai
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - E. T. Pásztor
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - M. Fagyas
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - J. Kalász
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - I. Édes
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - A. Tóth
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| | - Z. Papp
- Division of Clinical Physiology Institute of Cardiology Research Center for Molecular Medicine Faculty of Medicine University of Debrecen Debrecen Hungary
| |
Collapse
|
252
|
Ghadri JR, Sarcon A, Jaguszewski M, Diekmann J, Bataiosu RD, Hellermann J, Csordas A, Baumann L, Schöni AA, Lüscher TF, Templin C. Gender disparities in acute coronary syndrome. J Cardiovasc Med (Hagerstown) 2015; 16:355-62. [DOI: 10.2459/jcm.0000000000000248] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
253
|
Morgan PE, Laura RP, Maki RA, Reynolds WF, Davies MJ. Thiocyanate supplementation decreases atherosclerotic plaque in mice expressing human myeloperoxidase. Free Radic Res 2015; 49:743-9. [PMID: 25812586 DOI: 10.3109/10715762.2015.1019347] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Elevated levels of the heme enzyme myeloperoxidase (MPO) are associated with adverse cardiovascular outcomes. MPO predominantly catalyzes formation of the oxidants hypochlorous acid (HOCl) from Cl(-), and hypothiocyanous acid (HOSCN) from SCN(-), with these anions acting as competitive substrates. HOSCN is a less powerful and more specific oxidant than HOCl, and selectively targets thiols; such damage is largely reversible, unlike much HOCl-induced damage. We hypothesized that increased plasma SCN(-), and hence HOSCN formation instead of HOCl, may decrease artery wall damage. This was examined using high-fat fed atherosclerosis-prone LDLR(-/-) mice transgenic for human MPO, with and without SCN(-) (10 mM) added to drinking water. Serum samples, collected fortnightly, were analyzed for cholesterol, triglycerides, thiols, MPO, and SCN(-); study-long exposure was calculated by area under the curve (AUC). Mean serum SCN(-) concentrations were elevated in the supplemented mice (200-320 μM) relative to controls (< 120 μM). Normalized aortic root plaque areas at sacrifice were 26% lower in the SCN(-)-supplemented mice compared with controls (P = 0.0417), but plaque morphology was not appreciably altered. Serum MPO levels steadily increased in mice on the high-fat diet, however, comparison of SCN(-)-supplemented versus control mice showed no significant changes in MPO protein, cholesterol, or triglyceride levels; thiol levels were decreased in supplemented mice at one time-point. Plaque areas increased with higher cholesterol AUC (r = 0.4742; P = 0.0468), and decreased with increasing SCN(-) AUC (r = - 0.5693; P = 0.0134). These data suggest that increased serum SCN(-) levels, which can be achieved in humans by dietary manipulation, may decrease atherosclerosis burden.
Collapse
Affiliation(s)
- P E Morgan
- The Heart Research Institute , Newtown, NSW , Australia
| | | | | | | | | |
Collapse
|
254
|
Oxidative stress: dual pathway induction in cardiorenal syndrome type 1 pathogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:391790. [PMID: 25821554 PMCID: PMC4364374 DOI: 10.1155/2015/391790] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/11/2015] [Accepted: 02/15/2015] [Indexed: 01/07/2023]
Abstract
Cardiorenal Syndrome Type 1 (Type 1) is a specific condition which is characterized by a rapid worsening of cardiac function leading to acute kidney injury (AKI). Even though its pathophysiology is complex and not still completely understood, oxidative stress seems to play a pivotal role. In this study, we examined the putative role of oxidative stress in the pathogenesis of CRS Type 1. Twenty-three patients with acute heart failure (AHF) were included in the study. Subsequently, 11 patients who developed AKI due to AHF were classified as CRS Type 1. Quantitative determinations for IL-6, myeloperoxidase (MPO), nitric oxide (NO), copper/zinc superoxide dismutase (Cu/ZnSOD), and endogenous peroxidase activity (EPA) were performed. CRS Type 1 patients displayed significant augmentation in circulating ROS and RNS, as well as expression of IL-6. Quantitative analysis of all oxidative stress markers showed significantly lower oxidative stress levels in controls and AHF compared to CRS Type 1 patients (P < 0.05). This pilot study demonstrates the significantly heightened presence of dual oxidative stress pathway induction in CRS Type 1 compared to AHF patients. Our findings indicate that oxidative stress is a potential therapeutic target, as it promotes inflammation by ROS/RNS-linked pathogenesis.
Collapse
|
255
|
Liebetrau C, Hoffmann J, Dörr O, Gaede L, Blumenstein J, Biermann H, Pyttel L, Thiele P, Troidl C, Berkowitsch A, Rolf A, Voss S, Hamm CW, Nef H, Möllmann H. Release Kinetics of Inflammatory Biomarkers in a Clinical Model of Acute Myocardial Infarction. Circ Res 2015; 116:867-75. [DOI: 10.1161/circresaha.116.304653] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rationale:
Inflammation in the setting of acute myocardial infarction (MI) has been linked to risk stratification; however, the release kinetics of inflammatory biomarkers in patients with acute MI has been difficult to establish.
Objective:
The aim of this study was to determine the kinetics of changes in the levels of several biomarkers specifically linked to inflammation after transcoronary ablation of septal hypertrophy, a procedure that mimics acute MI.
Methods and Results:
We analyzed release kinetics of C-reactive protein, high-sensitivity C-reactive protein, interleukin-6, soluble CD40 ligand, and peripheral blood leukocyte subsets in patients (n=21) undergoing transcoronary ablation of septal hypertrophy. Blood samples were collected before transcoronary ablation of septal hypertrophy and at various times after transcoronary ablation of septal hypertrophy. Serum levels of C-reactive protein were increased at 24 hours (1.0 mg/dL [interquartile range [IQR], 0.7–1.75] versus 0.2 mg/dL [IQR, 0.1–1.05] at baseline [BL];
P
<0.001), whereas high-sensitivity C-reactive protein increased as early as 8 hours (2.68 mg/L [IQR, 1.23–11.80] versus 2.17 mg/L [IQR, 1.15–5.06] at BL;
P
=0.002). Interleukin-6 was significantly increased at 45 minutes (2.59 pg/mL [IQR, 1.69–5.0] versus 1.5 pg/mL [IQR, 1.5–2.21] at BL;
P
=0.002), and soluble CD40 ligand was significantly decreased at 60 minutes (801.6 pg/mL [IQR, 675.0–1653.5] versus 1750.0 pg/mL [IQR, 1151.0–2783.0] at BL;
P
=0.016). Elevated counts of polymorphonuclear neutrophils were detectable at 15 minutes, with a significant increase at 2 hours (6415 cells/μL [IQR, 5288–7827] versus 4697 cells/μL [IQR, 2892–5620] at BL;
P
=0.004). Significant monocytosis was observed at 24 hours (729 cells/μL [IQR, 584–1344] versus 523 cells/μL [IQR, 369–701] at BL;
P
=0.015).
Conclusions:
Interleukin-6 and neutrophil granulocytes showed a continuous rise at all prespecified time points after induction of MI. Our results provide valuable additional evidence of the diagnostic value of inflammatory biomarkers in the setting of early acute MI.
Collapse
Affiliation(s)
- Christoph Liebetrau
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Jedrzej Hoffmann
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Oliver Dörr
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Luise Gaede
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Johannes Blumenstein
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Hannes Biermann
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Lukas Pyttel
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Peter Thiele
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Christian Troidl
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Alexander Berkowitsch
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Andreas Rolf
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Sandra Voss
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Christian W. Hamm
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Holger Nef
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| | - Helge Möllmann
- From the Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); DZHK (German Centre for Cardiovascular Research), partner site Rheine-Main, Bad Nauheim, Germany (C.L., J.H., L.G., J.B., H.B., L.P., P.T., C.T., A.B., A.R., S.V., C.W.H., H.M.); and Department of Internal Medicine I, Division of Cardiology, University of Giessen, Giessen, Germany (C.L., O.D., C.W.H., H.N.)
| |
Collapse
|
256
|
Mehta N, Reilly M. Atherosclerotic Cardiovascular Disease Risk in the HAART-Treated HIV-1 Population. HIV CLINICAL TRIALS 2015; 6:5-24. [PMID: 15765307 DOI: 10.1310/ht0w-nx2n-u2bm-7luu] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Atherosclerotic cardiovascular disease (CVD), a leading cause of morbidity and mortality in the general population, is also an increasing cause for concern for HIV-infected patients. A number of risk factors for CVD are also associated with HIV disease and HIV therapy, particularly insulin resistance, metabolic dyslipidemia, and inflammation. For example, atherogenic dyslipidemia, a side effect of HIV therapy, is an established risk for CVD in the non-HIV-infected population. As our understanding of atherosclerotic disease evolves, new markers of CVD risk have been identified, including metabolic syndrome definitions and C-reactive protein, a marker of inflammation. Use of these markers, in association with established risk factor guidelines, may serve as important tools in helping HIV physicians implement drug regimens that allow optimum management of metabolic complications associated with HIV and HAART, and thereby reduce CVD risk. The objective of this article is to review the mechanisms of atherosclerotic CVD and to discuss risk factors and markers that can be applied in the evaluation and treatment of CVD in the HIV-positive population.
Collapse
Affiliation(s)
- Nehal Mehta
- Cardiovascular Division, Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
257
|
Gómez García A, Rivera Rodríguez M, Gómez Alonso C, Rodríguez Ochoa DY, Alvarez Aguilar C. Myeloperoxidase is associated with insulin resistance and inflammation in overweight subjects with first-degree relatives with type 2 diabetes mellitus. Diabetes Metab J 2015; 39:59-65. [PMID: 25729714 PMCID: PMC4342538 DOI: 10.4093/dmj.2015.39.1.59] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Family history of type 2 diabetes mellitus (T2DM) is one of risk factors for that in future a subject can develop diabetes. Insulin resistance (IR) is important in the pathogenesis of T2DM. There is evidence that oxidative stress plays an important role in the etiology and/or progression of diabetes. Myeloperoxidase (MPO) participates in developing of inflammation. The objective was to investigate if MPO is associated with IR and inflammation in individuals with first-degree relatives of T2DM. METHODS Cross-sectional study in 84 overweight individuals with family history of T2DM divided in two groups according to IR, group with IR (homeostasis model assessment [HOMA] ≥2.5; n=43) and control group (CG; HOMA <2.5; n=41). Complete clinical history and a venous blood sample were collected for measuring glucose and lipids profile, insulin, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), MPO, glutathione reductase (GRd), glutathione peroxidase, and superoxide dismutase. RESULTS MPO, TNF-α, and IL-6 were higher in patients with IR than in CG (MPO: 308.35 [190.85 to 445.42] vs. 177.35 [104.50 to 279.85], P=0.0001; TNF-α: 13.46 [10.58 to 18.88] vs. 9.39 [7.53 to 11.25], P=0.0001; IL-6: 32.93 [24.93 to 38.27] vs. 15.60 [12.93 to 26.27]; P=0.0001, respectively). MPO was associated with IR (rho de Spearman=0.362, P=0.001). In the analysis of lineal regression, MPO predicts IR (β, 0.263; t, 2.520; P=0.014). In the univariate analysis, MPO had an odds ratio of 9.880 for risk of IR (95% confidence interval, 2.647 to 36.879). CONCLUSION MPO had relation with IR and inflammation parameters in overweight subjects with first-degree relatives of T2DM. We need studies on a casual relationship and molecular mechanisms among the increased serum MPO levels, inflammation markers, and IR.
Collapse
Affiliation(s)
- Anel Gómez García
- Biomedical Research Center of Michoacán, Mexican Institute of Social Security, Morelia, Mexico
| | | | - Carlos Gómez Alonso
- Biomedical Research Center of Michoacán, Mexican Institute of Social Security, Morelia, Mexico
| | | | - Cleto Alvarez Aguilar
- Biomedical Research Center of Michoacán, Mexican Institute of Social Security, Morelia, Mexico
| |
Collapse
|
258
|
Najafi M, Mohammadi P. System study of MPO promoter high-frequency polymorphic variants on transcription factor network. Gene 2015; 560:143-8. [PMID: 25637720 DOI: 10.1016/j.gene.2015.01.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 01/22/2015] [Accepted: 01/27/2015] [Indexed: 11/19/2022]
Abstract
The neutrophil myeloperoxidase (MPO) promotes the oxidative stress by the production of active chlorinated molecules. The aim of this study was to investigate the association between MPO promoter polymorphic variants (rs2243827 and rs2333227) and, its serum level in patients with the stenosis of coronary arteries. Furthermore, a system approach was applied to create the MPO transcription factor network. A total of one hundred fifty six subjects (controls, stenosis<5%, n=71 and patients, stenosis>70%, n=85) undergoing coronary angiography were recruited. The polymorphic haplotypes and serum MPO level were identified using ARMS-PCR and ELISA techniques, respectively. The MPO transcription factor network was primarily created with PSICQUIC and ChIP data and, was improved with the predicted transcription factors. The regression analyses did not show an association between the serum MPO level and the extent of stenosis in coronary arteries. The network showed that the predicted transcription factors at the flanking regions of polymorphic variants are not directly interacted to MPO. In conclusion, the population and prediction studies showed no association between the serum MPO level, the promoter high-frequency polymorphic frequencies and the extent of stenosis in coronary arteries. A gene sub-cluster with MYB as central node was suggested to be involved with MPO on the transcription factor network.
Collapse
Affiliation(s)
- Mohammad Najafi
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
259
|
Abstract
The vasculature, composed of vessels of different morphology and function, distributes blood to all tissues and maintains physiological tissue homeostasis. In pathologies, the vasculature is often affected by, and engaged in, the disease process. This may result in excessive formation of new, unstable, and hyperpermeable vessels with poor blood flow, which further promotes hypoxia and disease propagation. Chronic vessel permeability may also facilitate metastatic spread of cancer. Thus, there is a strong incentive to learn more about an important aspect of vessel biology in health and disease: the regulation of vessel permeability. The current review aims to summarize current insights into different mechanisms of vascular permeability, its regulatory factors, and the consequences for disease.
Collapse
Affiliation(s)
- Lena Claesson-Welsh
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|
260
|
Zhang Y, Liu J, Li S, Xu RX, Sun J, Tang Y, Li JJ. Proprotein convertase subtilisin/kexin type 9 expression is transiently up-regulated in the acute period of myocardial infarction in rat. BMC Cardiovasc Disord 2014; 14:192. [PMID: 25519174 PMCID: PMC4279995 DOI: 10.1186/1471-2261-14-192] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/11/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The proprotein convertase subtilisin/kexin type 9 (PCSK9) has been confirmed as a major factor regulating cholesterol homeostasis and has low-density lipoprotein receptor (LDLR) independent effects. In addition, the pathogenesis of acute myocardial infarction (AMI) involves lipids alteration and other acute phase responses. It remains unknown whether the PCSK9 expression is influenced by the impact of AMI. The present study aimed to investigate the changes of PCSK9 concentration using AMI rat model. METHODS AMI (n = 6-8 at each time point) or sham operated (n = 6) adult male rats model were used. Whole blood and liver tissue were collected at 1, 3, 6, 9, 12, 24, 48, and 96 hour (h) post infarction. The plasma PCSK9 concentration was measured by ELISA and lipid profiles were measured by enzymatic assay. The liver mRNA levels of PCSK9, LDLR, sterol response element binding protein-2 (SREBP-2) and hepatocyte nuclear factor 1α (HNF1α) were measured by quantitative real-time PCR. RESULTS The plasma PCSK9 concentration was increased from 12 h to 96 h (P < 0.05 vs. control). Paralleled with the enhanced plasma PCSK9 concentration, the hepatic PCSK9 mRNA expression was up-regulated by 2.2-fold at 12 h and 4.1-fold at 24 h. Hepatic mRNA levels of LDLR, SREBP-2 and HNF1α were all increased and lipid profiles underwent great changes at this acute period. CONCLUSIONS We firstly demonstrated that PCSK9 was transiently up-regulated in the acute period of AMI, which is also driven by transcriptional factors, SREBP-2 and HNF1α, suggesting that the role of PCSK9 in myocardial injury may be needed further study.
Collapse
Affiliation(s)
| | | | | | | | | | - Yue Tang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing 100037, China.
| | | |
Collapse
|
261
|
Koch C, Henrich M, Heidt MC. Sequential analysis of myeloperoxidase for prediction of adverse events after suspected acute coronary ischemia. Clin Cardiol 2014; 37:744-9. [PMID: 25403739 DOI: 10.1002/clc.22336] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/18/2014] [Accepted: 08/20/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Myeloperoxidase (MPO) plasma values predict major adverse cardiac events (MACE) in cases of acute coronary syndrome. The effect of serial testing in patients who are suspected for acute coronary ischemia is unclear. HYPOTHESIS We hypothesize that sequential MPO measurement may improve prediction of MACE in patients with suspected acute coronary ischemia. METHODS The present prospective observational study examined the prognostic significance of MPO in 917 patients with suspicion of acute coronary syndrome. Blood samples were taken at cardiac catheter laboratory admission and the day after coronary angiography. We recorded patients' mortality, the occurrence of cardiac ischemia, and repeated percutaneous coronary intervention through the next 6 months. RESULTS Mortality among patients with increased MPO plasma levels the day after coronary angiography was increased significantly (P < 0.01). Patients with MPO values above 306.3 pmol/L had a significantly higher incidence of 6-month MACE (P < 0.0001) than patients with lower plasma values. Cox proportional hazards multivariate regression analyses revealed that MPO was an independent marker for MACE after suspected acute coronary ischemia (P = 0.048). However, MPO plasma levels at cardiac catheter laboratory admission showed no prognostic significance. CONCLUSIONS In patients with suspected myocardial infarction, MPO levels above 306.3 pmol/L measured 24 hours after onset of symptoms were independent predictors of 6-month mortality and MACE.
Collapse
Affiliation(s)
- Christian Koch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Giessen and Marburg, Giessen, Germany
| | | | | |
Collapse
|
262
|
Myeloperoxidase oxidized LDL interferes with endothelial cell motility through miR-22 and heme oxygenase 1 induction: possible involvement in reendothelialization of vascular injuries. Mediators Inflamm 2014; 2014:134635. [PMID: 25530680 PMCID: PMC4233670 DOI: 10.1155/2014/134635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/09/2014] [Indexed: 01/19/2023] Open
Abstract
Cardiovascular disease linked to atherosclerosis is the leading cause of death worldwide. Atherosclerosis is mainly linked to dysfunction in vascular endothelial cells and subendothelial accumulation of oxidized forms of LDL. In the present study, we investigated the role of myeloperoxidase oxidized LDL (Mox-LDL) in endothelial cell dysfunction. We studied the effect of proinflammatory Mox-LDL treatment on endothelial cell motility, a parameter essential for normal vascular processes such as angiogenesis and blood vessel repair. This is particularly important in the context of an atheroma plaque, where vascular wall integrity is affected and interference with its repair could contribute to progression of the disease. We investigated in vitro the effect of Mox-LDL on endothelial cells angiogenic properties and we also studied the signalling pathways that could be affected by analysing Mox-LDL effect on the expression of angiogenesis-related genes. We report that Mox-LDL inhibits endothelial cell motility and tubulogenesis through an increase in miR-22 and heme oxygenase 1 expression. Our in vitro data indicate that Mox-LDL interferes with parameters associated with angiogenesis. They suggest that high LDL levels in patients would impair their endothelial cell capacity to cope with a damaged endothelium contributing negatively to the progression of the atheroma plaque.
Collapse
|
263
|
Increased A3AR-dependent Vasoconstriction in Diabetic Mice Is Promoted by Myeloperoxidase. J Cardiovasc Pharmacol 2014; 64:465-72. [DOI: 10.1097/fjc.0000000000000139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
264
|
Tirotta I, Dichiarante V, Pigliacelli C, Cavallo G, Terraneo G, Bombelli FB, Metrangolo P, Resnati G. (19)F magnetic resonance imaging (MRI): from design of materials to clinical applications. Chem Rev 2014; 115:1106-29. [PMID: 25329814 DOI: 10.1021/cr500286d] [Citation(s) in RCA: 340] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ilaria Tirotta
- Laboratory of Nanostructured Fluorinated Materials (NFMLab), Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta" and ‡Fondazione Centro Europeo Nanomedicina, Politecnico di Milano , Milan 20131, Italy
| | | | | | | | | | | | | | | |
Collapse
|
265
|
Misztal T, Rusak T, Tomasiak M. Clinically relevant HOCl concentrations reduce clot retraction rate via the inhibition of energy production in platelet mitochondria. Free Radic Res 2014; 48:1443-53. [DOI: 10.3109/10715762.2014.960866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
266
|
Mayyas FA, Al-Jarrah MI, Ibrahim KS, Alzoubi KH. Level and significance of plasma myeloperoxidase and the neutrophil to lymphocyte ratio in patients with coronary artery disease. Exp Ther Med 2014; 8:1951-1957. [PMID: 25371762 PMCID: PMC4218701 DOI: 10.3892/etm.2014.2034] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 10/06/2014] [Indexed: 12/14/2022] Open
Abstract
Inflammation plays a pivotal role in the etiology of coronary artery disease (CAD). Myeloperoxidase (MPO) is a potent inflammatory factor and a critical modulator of coronary inflammation and oxidative stress. The goal of this study was to determine the impact of the plasma MPO (pMPO) level and neutrophil/lymphocyte ratio on the clinical characteristics and outcomes of patients with CAD. Blood samples were collected from 210 patients with underlying chest pain or recent myocardial infarction (MI) prior to coronary angiography in order to measure pMPO levels. The pMPO levels and neutrophil/lymphocyte ratio were correlated with clinical characteristics and outcomes following catheterization. The pMPO level and neutrophil/lymphocyte ratio were higher in patients with recent MI than in patients with CAD (coronary occlusion ≥50%) or without CAD (coronary occlusion <50%). Patients with ST segment elevated MI (STEMI) had a higher neutrophil/lymphocyte ratio relative to patients with non-STEMI. The pMPO level was identified to correlate with the neutrophil/lymphocyte ratio and the need for coronary artery reperfusion by coronary artery bypass surgery or percutaneous coronary intervention. Patients who were taking aspirin had lower pMPO levels and neutrophil/lymphocyte ratio compared with those who were not taking aspirin. The plasma neutrophil/lymphocyte ratio was negatively associated with the left ventricular ejection fraction at baseline and the 30-day follow-up, whereas pMPO showed no correlation. Multivariate analysis indicated that the pMPO level was positively associated with MI, the neutrophil/lymphocyte ratio and coronary intervention. The preoperative use of aspirin was associated with a lower pMPO level and neutrophil/lymphocyte ratio. In conclusion, pMPO is positively associated with MI, the neutrophil/lymphocyte ratio and coronary intervention. The preoperative use of aspirin is associated with a lower pMPO level and neutrophil/lymphocyte ratio. pMPO may serve as a predictor of coronary intervention and as a potential therapeutic target for the reduction of inflammation in patients with CAD.
Collapse
Affiliation(s)
- Fadia A Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad I Al-Jarrah
- Department of General Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan ; Princess Muna Heart Institute, King Abdullah University Hospital, Irbid 22110, Jordan
| | - Khalid S Ibrahim
- Princess Muna Heart Institute, King Abdullah University Hospital, Irbid 22110, Jordan ; Department of General Surgery, Division of Cardiovascular Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
267
|
Tian S, Hirshfield KM, Jabbour SK, Toppmeyer D, Haffty BG, Khan AJ, Goyal S. Serum biomarkers for the detection of cardiac toxicity after chemotherapy and radiation therapy in breast cancer patients. Front Oncol 2014; 4:277. [PMID: 25346912 PMCID: PMC4191171 DOI: 10.3389/fonc.2014.00277] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/23/2014] [Indexed: 12/31/2022] Open
Abstract
Multi-modality cancer treatments that include chemotherapy, radiation therapy, and targeted agents are highly effective therapies. Their use, especially in combination, is limited by the risk of significant cardiac toxicity. The current paradigm for minimizing cardiac morbidity, based on serial cardiac function monitoring, is suboptimal. An alternative approach based on biomarker testing, has emerged as a promising adjunct and a potential substitute to routine echocardiography. Biomarkers, most prominently cardiac troponins and natriuretic peptides, have been evaluated for their ability to describe the risk of potential cardiac dysfunction in clinically asymptomatic patients. Early rises in cardiac troponin concentrations have consistently predicted the risk and severity of significant cardiac events in patients treated with anthracycline-based chemotherapy. Biomarkers represent a novel, efficient, and robust clinical decision tool for the management of cancer therapy-induced cardiotoxicity. This article aims to review the clinical evidence that supports the use of established biomarkers such as cardiac troponins and natriuretic peptides, as well as emerging data on proposed biomarkers.
Collapse
Affiliation(s)
- Sibo Tian
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Kim M Hirshfield
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Deborah Toppmeyer
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Bruce G Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Atif J Khan
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Sharad Goyal
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| |
Collapse
|
268
|
Arbel Y, Shacham Y, Ziv-Baran T, Laufer Perl M, Finkelstein A, Halkin A, Revivo M, Milwidsky A, Berliner S, Herz I, Keren G, Banai S. Higher Neutrophil/Lymphocyte Ratio Is Related to Lower Ejection Fraction and Higher Long-term All-Cause Mortality in ST-Elevation Myocardial Infarction Patients. Can J Cardiol 2014; 30:1177-82. [DOI: 10.1016/j.cjca.2014.05.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/12/2014] [Accepted: 05/12/2014] [Indexed: 11/26/2022] Open
|
269
|
Panasenko OM, Gorudko IV, Sokolov AV. Hypochlorous acid as a precursor of free radicals in living systems. BIOCHEMISTRY (MOSCOW) 2014; 78:1466-89. [PMID: 24490735 DOI: 10.1134/s0006297913130075] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypochlorous acid (HOCl) is produced in the human body by the family of mammalian heme peroxidases, mainly by myeloperoxidase, which is secreted by neutrophils and monocytes at sites of inflammation. This review discusses the reactions that occur between HOCl and the major classes of biologically important molecules (amino acids, proteins, nucleotides, nucleic acids, carbohydrates, lipids, and inorganic substances) to form free radicals. The generation of such free radical intermediates by HOCl and other reactive halogen species is accompanied by the development of halogenative stress, which causes a number of socially important diseases, such as cardiovascular, neurodegenerative, infectious, and other diseases usually associated with inflammatory response and characterized by the appearance of biomarkers of myeloperoxidase and halogenative stress. Investigations aimed at elucidating the mechanisms regulating the activity of enzyme systems that are responsible for the production of reactive halogen species are a crucial step in opening possibilities for control of the development of the body's inflammatory response.
Collapse
Affiliation(s)
- O M Panasenko
- Research Institute of Physico-Chemical Medicine, Moscow, 119435, Russia.
| | | | | |
Collapse
|
270
|
Ikenaga H, Kurisu S, Watanabe N, Shimonaga T, Higaki T, Iwasaki T, Utsunomiya H, Mitsuba N, Ishibashi K, Dohi Y, Fukuda Y, Imai K, Sueda T, Kihara Y. Predictive value of neutrophil to lymphocyte ratio for the presence of coronary artery ectasia in patients with aortic aneurysms. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VESSELS 2014; 4:30-34. [PMID: 29450182 PMCID: PMC5801477 DOI: 10.1016/j.ijchv.2014.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 08/05/2014] [Accepted: 08/10/2014] [Indexed: 12/13/2022]
Abstract
Background Both aortic aneurysms and coronary artery ectasia (CAE) frequently coexist and are associated with more pronounced inflammation. Neutrophil to lymphocyte ratio (NL ratio) is widely used as a marker of inflammation. However, relation between CAE and NL ratio in patients with aortic aneurysms is not fully understood. This study was undertaken to assess relation between CAE and NL ratio in patients with aortic aneurysms. Methods This study consisted of 93 consecutive patients with aortic aneurysms (AA group) and 79 patients without aortic aneurysms who had angiographically normal coronary arteries as the control group. Moreover, patients with aortic aneurysms were classified into two groups based on the presence of CAE; CAE (+) group (n = 44) and CAE (−) group (n = 49). We compared blood chemical parameters in both groups. Results In the AA group, 44 patients (47.3%) had CAE. The AA group had a significantly higher NL ratio than the control group (2.93 ± 1.43 vs. 2.45 ± 1.05, p = 0.027). Furthermore, the CAE (+) group had a significantly higher NL ratio than the CAE (−) group (3.39 ± 1.67 vs. 2.52 ± 1.04, p < 0.01). Multivariate logistic regression analysis revealed that the high NL ratio was an independent predictor for CAE in patients with aortic aneurysms (odds ratio 1.76, 95% confidence interval 1.24–2.69, p = 0.001). Conclusions Patients with aortic aneurysms had a significantly higher NL ratio than those without aortic aneurysms. Furthermore, the NL ratio might predict the presence of CAE in patients with aortic aneurysms.
Collapse
Affiliation(s)
- Hiroki Ikenaga
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Satoshi Kurisu
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Noriaki Watanabe
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takashi Shimonaga
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tadanao Higaki
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Toshitaka Iwasaki
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Hiroto Utsunomiya
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Naoya Mitsuba
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Ken Ishibashi
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoshihiro Dohi
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yukihiro Fukuda
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Katsuhiko Imai
- Department of Cardiovascular Surgery, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Taijiro Sueda
- Department of Cardiovascular Surgery, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
271
|
G-CSF induces membrane expression of a myeloperoxidase glycovariant that operates as an E-selectin ligand on human myeloid cells. Proc Natl Acad Sci U S A 2014; 111:10696-701. [PMID: 25002508 DOI: 10.1073/pnas.1320833111] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The host defense response critically depends on the production of leukocytes by the marrow and the controlled delivery of these cells to relevant sites of inflammation/infection. The cytokine granulocyte-colony stimulating factor (G-CSF) is commonly used therapeutically to augment neutrophil recovery following chemo/radiation therapy for malignancy, thereby decreasing infection risk. Although best known as a potent inducer of myelopoiesis, we previously reported that G-CSF also promotes the delivery of leukocytes to sites of inflammation by stimulating expression of potent E-selectin ligands, including an uncharacterized ∼65-kDa glycoprotein. To identify this ligand, we performed integrated biochemical analysis and mass spectrometry studies of G-CSF-treated primary human myeloid cells. Our studies show that this novel E-selectin ligand is a glycoform of the heavy chain component of the enzyme myeloperoxidase (MPO), a well-known lysosomal peroxidase. This specialized MPO glycovariant, referred to as "MPO-E-selectin ligand" (MPO-EL), is expressed on circulating G-CSF-mobilized leukocytes and is naturally expressed on blood myeloid cells in patients with febrile leukocytosis. In vitro biochemical studies show that G-CSF programs MPO-EL expression on human blood leukocytes and marrow myeloid cells via induction of N-linked sialofucosylations on MPO, with concomitant cell surface display of the molecule. MPO-EL is catalytically active and mediates angiotoxicity on human endothelial cells that express E-selectin. These findings thus define a G-CSF effect on MPO chemical biology that endows unsuspected functional versatility upon this enzyme, unveiling new perspectives on the biology of G-CSF and MPO, and on the role of E-selectin receptor/ligand interactions in leukocyte migration and vascular pathology.
Collapse
|
272
|
Carbon nanotube wiring for signal amplification of electrochemical magneto immunosensors: application to myeloperoxidase detection. Anal Bioanal Chem 2014; 406:5487-93. [DOI: 10.1007/s00216-014-7954-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/30/2014] [Accepted: 06/05/2014] [Indexed: 01/30/2023]
|
273
|
Sen SK, Boelte KC, Barb JJ, Joehanes R, Zhao X, Cheng Q, Adams L, Teer JK, Accame DS, Chowdhury S, Singh LN, Kavousi M, Peyser PA, Quigley L, Priel DL, Lau K, Kuhns DB, Yoshimura T, Johnson AD, Hwang SJ, Chen MY, Arai AE, Green ED, Mullikin JC, Kolodgie FD, O'Donnell CJ, Virmani R, Munson PJ, McVicar DW, Biesecker LG. Integrative DNA, RNA, and protein evidence connects TREML4 to coronary artery calcification. Am J Hum Genet 2014; 95:66-76. [PMID: 24975946 PMCID: PMC4085627 DOI: 10.1016/j.ajhg.2014.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 06/04/2014] [Indexed: 12/18/2022] Open
Abstract
Coronary artery calcification (CAC) is a heritable and definitive morphologic marker of atherosclerosis that strongly predicts risk for future cardiovascular events. To search for genes involved in CAC, we used an integrative transcriptomic, genomic, and protein expression strategy by using next-generation DNA sequencing in the discovery phase with follow-up studies using traditional molecular biology and histopathology techniques. RNA sequencing of peripheral blood from a discovery set of CAC cases and controls was used to identify dysregulated genes, which were validated by ClinSeq and Framingham Heart Study data. Only a single gene, TREML4, was upregulated in CAC cases in both studies. Further examination showed that rs2803496 was a TREML4 cis-eQTL and that the minor allele at this locus conferred up to a 6.5-fold increased relative risk of CAC. We characterized human TREML4 and demonstrated by immunohistochemical techniques that it is localized in macrophages surrounding the necrotic core of coronary plaques complicated by calcification (but not in arteries with less advanced disease). Finally, we determined by von Kossa staining that TREML4 colocalizes with areas of microcalcification within coronary plaques. Overall, we present integrative RNA, DNA, and protein evidence implicating TREML4 in coronary artery calcification. Our findings connect multimodal genomics data with a commonly used clinical marker of cardiovascular disease.
Collapse
Affiliation(s)
- Shurjo K Sen
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | | | - Jennifer J Barb
- Center for Information Technology, NIH, Bethesda, MD 20892, USA
| | - Roby Joehanes
- Center for Information Technology, NIH, Bethesda, MD 20892, USA
| | | | - Qi Cheng
- CVPath Institute, Gaithersburg, MD 20878, USA
| | - Lila Adams
- CVPath Institute, Gaithersburg, MD 20878, USA
| | | | - David S Accame
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Soma Chowdhury
- Center for Biologics Evaluation and Research, FDA, Bethesda, MD 20892, USA
| | - Larry N Singh
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Maryam Kavousi
- Netherlands Genomics-Initiative-Sponsored Netherlands Consortium for Healthy Aging and Department of Epidemiology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48104, USA
| | - Laura Quigley
- National Cancer Institute, NIH, Frederick, MD 21702, USA
| | - Debra Long Priel
- Applied/Developmental Research Directorate, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Karen Lau
- Applied/Developmental Research Directorate, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Douglas B Kuhns
- Applied/Developmental Research Directorate, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | | | - Andrew D Johnson
- Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA; National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
| | - Shih-Jen Hwang
- Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA; National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
| | - Marcus Y Chen
- Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Andrew E Arai
- Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Eric D Green
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - James C Mullikin
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | | | - Christopher J O'Donnell
- Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA; National Heart, Lung and Blood Institute's Framingham Heart Study, Framingham, MA 01702, USA
| | | | - Peter J Munson
- Center for Information Technology, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
274
|
Lehners A, Lange S, Niemann G, Rosendahl A, Meyer-Schwesinger C, Oh J, Stahl R, Ehmke H, Benndorf R, Klinke A, Baldus S, Wenzel UO. Myeloperoxidase deficiency ameliorates progression of chronic kidney disease in mice. Am J Physiol Renal Physiol 2014; 307:F407-17. [PMID: 24990898 DOI: 10.1152/ajprenal.00262.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Myeloperoxidase (MPO) is an enzyme expressed in neutrophils and monocytes/macrophages. Beside its well-defined role in innate immune defence, it may also be responsible for tissue damage. To identify the role of MPO in the progression of chronic kidney disease (CKD), we investigated CKD in a model of renal ablation in MPO knockout and wild-type mice. CKD was induced by 5/6 nephrectomy. Mice were followed for 10 wk to evaluate the impact of MPO deficiency on renal morbidity. Renal ablation induced CKD in wild-type mice with increased plasma levels of MPO compared with controls. No difference was found between MPO-deficient and wild-type mice regarding albuminuria 1 wk after renal ablation, indicating similar acute responses to renal ablation. Over the next 10 wk, however, MPO-deficient mice developed significantly less albuminuria and glomerular injury than wild-type mice. This was accompanied by a significantly lower renal mRNA expression of the fibrosis marker genes plasminogen activator inhibitor-I, collagen type III, and collagen type IV as well as matrix metalloproteinase-2 and matrix metalloproteinase-9. MPO-deficient mice also developed less renal inflammation after renal ablation, as indicated by a lower infiltration of CD3-positive T cells and F4/80-positive monocytes/macrophages compared with wild-type mice. In vitro chemotaxis of monocyte/macrophages isolated from MPO-deficient mice was impaired compared with wild-type mice. No significant differences were observed for mortality and blood pressure after renal ablation. In conclusion, these results demonstrate that MPO deficiency ameliorates renal injury in the renal ablation model of CKD in mice.
Collapse
Affiliation(s)
- Alexander Lehners
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sascha Lange
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Gianina Niemann
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Alva Rosendahl
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jun Oh
- Division of Nephrology, Department of Pediatrics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf Stahl
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Heimo Ehmke
- Department of Physiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Benndorf
- Clinical Pharmacology, University of Halle, Halle, Germany; and
| | - Anna Klinke
- Division of Cardiology, University of Cologne, Cologne, Germany
| | - Stephan Baldus
- Division of Cardiology, University of Cologne, Cologne, Germany
| | - Ulrich Otto Wenzel
- Division of Nephrology, Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany;
| |
Collapse
|
275
|
|
276
|
Adam M, Gajdova S, Kolarova H, Kubala L, Lau D, Geisler A, Ravekes T, Rudolph V, Tsao PS, Blankenberg S, Baldus S, Klinke A. Red blood cells serve as intravascular carriers of myeloperoxidase. J Mol Cell Cardiol 2014; 74:353-63. [PMID: 24976018 DOI: 10.1016/j.yjmcc.2014.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 05/23/2014] [Accepted: 06/18/2014] [Indexed: 12/13/2022]
Abstract
Myeloperoxidase (MPO) is a heme enzyme abundantly expressed in polymorphonuclear neutrophils. MPO is enzymatically capable of catalyzing the generation of reactive oxygen species (ROS) and the consumption of nitric oxide (NO). Thus MPO has both potent microbicidal and, upon binding to the vessel wall, pro-inflammatory properties. Interestingly, MPO - a highly cationic protein - has been shown to bind to both endothelial cells and leukocyte membranes. Given the anionic surface charge of red blood cells, we investigated binding of MPO to erythrocytes. Red blood cells (RBCs) derived from patients with elevated MPO plasma levels showed significantly higher amounts of MPO by flow cytometry and ELISA than healthy controls. Heparin-induced MPO-release from patient-derived RBCs was significantly increased compared to controls. Ex vivo experiments revealed dose and time dependency for MPO-RBC binding, and immunofluorescence staining as well as confocal microscopy localized MPO-RBC interaction to the erythrocyte plasma membrane. NO-consumption by RBC-membrane fragments (erythrocyte "ghosts") increased with incrementally greater concentrations of MPO during incubation, indicating preserved catalytic MPO activity. In vivo infusion of MPO-loaded RBCs into C57BL/6J mice increased local MPO tissue concentrations in liver, spleen, lung, and heart tissue as well as within the cardiac vasculature. Further, NO-dependent relaxation of aortic rings was altered by RBC bound-MPO and systemic vascular resistance significantly increased after infusion of MPO-loaded RBCs into mice. In summary, we find that MPO binds to RBC membranes in vitro and in vivo, is transported by RBCs to remote sites in mice, and affects endothelial function as well as systemic vascular resistance. RBCs may avidly bind circulating MPO, and act as carriers of this leukocyte-derived enzyme.
Collapse
Affiliation(s)
- Matti Adam
- Stanford University, Division of Cardiovascular Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford, CA, USA.
| | - Silvie Gajdova
- Academy of Sciences of the Czech Republic, Institute of Biophysics, Brno, Czech Republic
| | - Hana Kolarova
- Academy of Sciences of the Czech Republic, Institute of Biophysics, Brno, Czech Republic
| | - Lukas Kubala
- Academy of Sciences of the Czech Republic, Institute of Biophysics, Brno, Czech Republic; International Clinical Research Center-CBCE, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Denise Lau
- University of Hamburg, Heart Center, Department of Cardiovascular Medicine, Hamburg, Germany
| | - Anne Geisler
- University of Hamburg, Heart Center, Department of Cardiovascular Medicine, Hamburg, Germany
| | - Thorben Ravekes
- Heart Center, Department of Cardiology and Cologne Cardiovascular Research Center, University of Cologne, Cologne, Germany
| | - Volker Rudolph
- Heart Center, Department of Cardiology and Cologne Cardiovascular Research Center, University of Cologne, Cologne, Germany
| | - Philip S Tsao
- Stanford University, Division of Cardiovascular Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Stefan Blankenberg
- University of Hamburg, Heart Center, Department of Cardiovascular Medicine, Hamburg, Germany
| | - Stephan Baldus
- Heart Center, Department of Cardiology and Cologne Cardiovascular Research Center, University of Cologne, Cologne, Germany
| | - Anna Klinke
- Heart Center, Department of Cardiology and Cologne Cardiovascular Research Center, University of Cologne, Cologne, Germany
| |
Collapse
|
277
|
Doura T, Hata R, Nonaka H, Sugihara F, Yoshioka Y, Sando S. An adhesive (19)F MRI chemical probe allows signal off-to-on-type molecular sensing in a biological environment. Chem Commun (Camb) 2014; 49:11421-3. [PMID: 24169571 DOI: 10.1039/c3cc46471g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We report a new strategy for designing a signal off-to-on-type (19)F MRI chemical probe that operates in biological environments. The present strategy is based on the control of adherence of a (19)F MRI chemical probe to certain blood proteins, accompanied by a change in transverse relaxation time of (19)F nuclei.
Collapse
Affiliation(s)
- Tomohiro Doura
- INAMORI Frontier Research Center, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
278
|
Searle J, Shih J, Muller R, Vollert JO, Müller C, Danne O, Datwyler S, Möckel M. The role of myeloperoxidase (MPO) for prognostic evaluation in sensitive cardiac troponin I negative chest pain patients in the emergency department. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2014; 2:203-10. [PMID: 24222831 DOI: 10.1177/2048872613484688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/07/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND The diagnostic work-up of patients with acute chest pain in the emergency department (ED) is a challenging task. Serial troponin testing is required to rule-out acute myocardial infarction. OBJECTIVE To evaluate the value of myeloperoxidase (MPO) testing in sensitive cardiac troponin I (cTnI) negative patients with suspected acute coronary syndromes (ACS) in the routine setting of an ED. METHODS MPO was assessed in 432 consecutive patients presenting to the ED with ACS. In 266 patients, serial blood samples were available. After 6 weeks, major adverse cardiac events (MACE) were assessed. MPO and cTnI were measured in all available samples. For cTnI, a sensitive assay was used. Cut-off values were derived from an independent sample of 300 healthy volunteers. RESULTS Incidence of MACE in our population was 13%. MPO levels revealed sensitivity (Sens) of 82.1% and specificity (Spec) of 37.2% for MACE compared with 60.7% Sens and 61.4% Spec for sensitive cTnI. In serial sensitive cTnI negative patients (n=218), MACE incidence was 6.4%. MPO continued to demonstrate significant discriminatory power for the prognosis of MACE. Multivariate analyses confirmed these findings. CONCLUSION MPO has an independent prognostic value overall and most notably in patients tested negative with a higher sensitive cardiac troponin I assay. MPO could be a promising biomarker for the initial evaluation of patients in chest pain units and is worth further investigation.
Collapse
Affiliation(s)
- Julia Searle
- Department of Cardiology, CVK and Emergency Medicine CVK, CCM, Charité - Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
279
|
Krintus M, Kozinski M, Kubica J, Sypniewska G. Critical appraisal of inflammatory markers in cardiovascular risk stratification. Crit Rev Clin Lab Sci 2014; 51:263-79. [DOI: 10.3109/10408363.2014.913549] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
280
|
Ganji SH, Kamanna VS, Kashyap ML. Niacin decreases leukocyte myeloperoxidase: mechanistic role of redox agents and Src/p38MAP kinase. Atherosclerosis 2014; 235:554-61. [PMID: 24956528 DOI: 10.1016/j.atherosclerosis.2014.05.948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/15/2014] [Accepted: 05/24/2014] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Leukocyte myeloperoxidase (MPO) is a major player in the pathogenesis of various chronic diseases including atherosclerosis. This study proposes the novel concept that niacin, through reactive oxygen species (ROS)-mediated signaling, decreases neutrophil MPO release and its activity, protects apolipoprotein-AI (apo-AI) modification and improves HDL function. METHODS Human blood leukocytes and leukocytic cell line HL-60 cells were treated with niacin, and stimulated with phorbol myristate acetate (PMA). Cellular and released MPO activity in the medium was measured by assessing chlorination of MPO-specific substrate. MPO protein release in the medium and apo-AI degradation was measured by Western blot analysis. Monocyte adhesion to human aortic primary endothelial cells was measured to assess biological function of HDL/apo-AI. RESULTS PMA significantly increased leukocyte MPO activity in both intracellular extract and medium. Niacin (0.25-0.5 mM) decreased PMA-induced MPO activity (cellular and released in the media). Niacin also decreased MPO protein mass in the medium without affecting its mRNA expression. Increased NADPH oxidase and ROS production by PMA were also significantly inhibited by niacin. Studies with specific inhibitors suggest that ROS-dependent Src and p38MAP kinase mediate decreased MPO activity by niacin. Niacin blocked apo-AI degradation, and apo-AI from niacin treated cells decreased monocyte adhesion to aortic endothelial cells. CONCLUSIONS These findings identify niacin as a potent inhibitor of leukocyte MPO release and MPO-mediated formation of dysfunctional HDL. Niacin and niacin-related chemical entities may form important therapeutic agents for MPO-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Shobha H Ganji
- Atherosclerosis Research Center, Department of Veterans Affairs Healthcare System, Long Beach, CA, USA; The University of California, Irvine, CA, USA
| | - Vaijinath S Kamanna
- Atherosclerosis Research Center, Department of Veterans Affairs Healthcare System, Long Beach, CA, USA; The University of California, Irvine, CA, USA.
| | - Moti L Kashyap
- Atherosclerosis Research Center, Department of Veterans Affairs Healthcare System, Long Beach, CA, USA; The University of California, Irvine, CA, USA.
| |
Collapse
|
281
|
Sigurdardottir G, Ekman AK, Ståhle M, Bivik C, Enerbäck C. Systemic treatment and narrowband ultraviolet B differentially affect cardiovascular risk markers in psoriasis. J Am Acad Dermatol 2014; 70:1067-75. [DOI: 10.1016/j.jaad.2013.12.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/20/2013] [Accepted: 12/21/2013] [Indexed: 12/12/2022]
|
282
|
Abstract
Myeloperoxidase (MPO) plays a central role in the innate immune system by generating leukocyte-derived oxidants to combat invading pathogens. These reactive intermediates have been increasingly recognized to be potentially deleterious, causing oxidative injury in inflammatory disease states such as cardiovascular disease. Recent evidence now suggests that circulating MPO can act as a clinical prognostic indicator for patients with cardiovascular disease.
Collapse
|
283
|
Millard RW, Tranter M. Complementary, alternative, and putative nontroponin biomarkers of acute coronary syndrome: new resources for future risk assessment calculators. ACTA ACUST UNITED AC 2014; 67:312-20. [PMID: 24774594 DOI: 10.1016/j.rec.2013.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 11/16/2022]
Abstract
Biomarkers, other than cardiac troponin, with potential sensitivity and selectivity that provide diagnostic and prognostic insights into the tissue-specific injury processes underlying acute coronary syndrome and their possible use in risk stratification algorithms are discussed. Such biomarkers may be useful as complementary or alternative to cardiac troponin (I or T) assays in early diagnosis of acute coronary syndrome, as well as for monitoring acute coronary syndrome progression and prognosis assessment. The information included in this article is based on a critical analysis of selected published biomedical literature accessible through the United States National Library of Medicine's MEDLINE-PubMed and Scopus search engines. The majority of articles cited in this review and perspective, except for a few historical publications as background, were published between January 2000 and December 2013.
Collapse
Affiliation(s)
- Ronald W Millard
- Department of Pharmacology & Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States.
| | - Michael Tranter
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| |
Collapse
|
284
|
Yu M, Zhou H, Zhao J, Xiao N, Roychowdhury S, Schmitt D, Hu B, Ransohoff RM, Harding CV, Hise AG, Hazen SL, DeFranco AL, Fox PL, Morton RE, Dicorleto PE, Febbraio M, Nagy LE, Smith JD, Wang JA, Li X. MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases. ACTA ACUST UNITED AC 2014; 211:887-907. [PMID: 24752299 PMCID: PMC4010914 DOI: 10.1084/jem.20131314] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
MyD88-dependent GM-CSF production by endothelial cells plays a role in the initiation of obesity-associated inflammation by promoting adipose macrophage recruitment and M1-like polarization. Low-grade systemic inflammation is often associated with metabolic syndrome, which plays a critical role in the development of the obesity-associated inflammatory diseases, including insulin resistance and atherosclerosis. Here, we investigate how Toll-like receptor–MyD88 signaling in myeloid and endothelial cells coordinately participates in the initiation and progression of high fat diet–induced systemic inflammation and metabolic inflammatory diseases. MyD88 deficiency in myeloid cells inhibits macrophage recruitment to adipose tissue and their switch to an M1-like phenotype. This is accompanied by substantially reduced diet-induced systemic inflammation, insulin resistance, and atherosclerosis. MyD88 deficiency in endothelial cells results in a moderate reduction in diet-induced adipose macrophage infiltration and M1 polarization, selective insulin sensitivity in adipose tissue, and amelioration of spontaneous atherosclerosis. Both in vivo and ex vivo studies suggest that MyD88-dependent GM-CSF production from the endothelial cells might play a critical role in the initiation of obesity-associated inflammation and development of atherosclerosis by priming the monocytes in the adipose and arterial tissues to differentiate into M1-like inflammatory macrophages. Collectively, these results implicate a critical MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases.
Collapse
Affiliation(s)
- Minjia Yu
- Department of Immunology, 2 Department of Cellular and Molecular Medicine, 3 Department of Pathobiology, 4 Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH 44195
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
285
|
Scharnagl H, Kleber ME, Genser B, Kickmaier S, Renner W, Weihrauch G, Grammer T, Rossmann C, Winkelmann BR, Boehm BO, Sattler W, März W, Malle E. Association of myeloperoxidase with total and cardiovascular mortality in individuals undergoing coronary angiography--the LURIC study. Int J Cardiol 2014; 174:96-105. [PMID: 24746542 PMCID: PMC4045190 DOI: 10.1016/j.ijcard.2014.03.168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/10/2014] [Accepted: 03/26/2014] [Indexed: 12/30/2022]
Abstract
Background The phagocytic enzyme myeloperoxidase (MPO) acts as a front-line defender against microorganisms. However, increased MPO levels have been found to be associated with complex and calcified atherosclerotic lesions and incident cardiovascular disease. Therefore, this study aimed to investigate a predictive role of MPO, a biomarker of inflammation and oxidative stress, for total and cardiovascular mortality in patients referred to coronary angiography. Methods and results MPO plasma concentrations along with eight MPO polymorphisms were determined in 3036 participants of the Ludwigshafen Risk and Cardiovascular Health study (median follow-up 7.75 years). MPO concentrations were positively associated with age, diabetes, smoking, markers of systemic inflammation (interleukin-6, fibrinogen, C-reactive protein, serum amyloid A) and vascular damage (vascular cellular adhesion molecule-1 and intercellular adhesion molecule-1) but negatively associated with HDL-cholesterol and apolipoprotein A-I. After adjustment for cardiovascular risk factors MPO concentrations in the highest versus the lowest quartile were associated with a 1.34-fold risk (95% CI: 1.09–1.67) for total mortality. In the adjusted model the hazard ratio for cardiovascular mortality in the highest MPO quartile was 1.42 (95% CI: 1.07–1.88). Five MPO polymorphisms were positively associated with MPO concentrations but not with mortality. Using Mendelian randomization, we did not obtain evidence for a causal association of MPO with either total or cardiovascular mortality. Conclusions MPO concentrations but not genetic variants at the MPO locus are independently associated with risk for total and cardiovascular mortality in coronary artery disease patients.
Collapse
Affiliation(s)
- Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Marcus E Kleber
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bernd Genser
- Mannheim Institute of Public Health, Medical Faculty Mannheim, Rupertus Carola University of Heidelberg, Mannheim, Germany
| | - Sandra Kickmaier
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Austria
| | - Wilfried Renner
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Gisela Weihrauch
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Tanja Grammer
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christine Rossmann
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Austria
| | | | - Bernhard O Boehm
- Ulm University Medical Centre, Department of Internal Medicine I, Ulm University, Ulm, Germany; LKC School of Medicine, Nanyang Technological University, Singapore and Imperial College London, UK
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Austria
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria; Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Synlab Academy, Mannheim, Germany
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Austria.
| |
Collapse
|
286
|
Prognostic Performance of Multiple Biomarkers in Patients With Non–ST-Segment Elevation Acute Coronary Syndrome. J Am Coll Cardiol 2014; 63:1644-53. [DOI: 10.1016/j.jacc.2013.12.034] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 10/20/2013] [Accepted: 12/17/2013] [Indexed: 12/19/2022]
|
287
|
Millard RW, Tranter M. Biomarcadores no troponínicos, complementarios, alternativos y presuntos, para el síndrome coronario agudo: nuevos recursos para los futuros instrumentos de cálculo del riesgo. Rev Esp Cardiol 2014. [DOI: 10.1016/j.recesp.2013.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
288
|
Sokolov AV, Kostevich VA, Runova OL, Gorudko IV, Vasilyev VB, Cherenkevich SN, Panasenko OM. Proatherogenic modification of LDL by surface-bound myeloperoxidase. Chem Phys Lipids 2014; 180:72-80. [PMID: 24631066 DOI: 10.1016/j.chemphyslip.2014.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 01/31/2014] [Accepted: 02/24/2014] [Indexed: 01/28/2023]
Abstract
One of the factors promoting oxidative/halogenating modification of low-density lipoproteins (LDL) is myeloperoxidase (MPO). We have shown previously that MPO binds to the LDL surfaces. The LDL-MPO complex is uncoupled in the presence of peptide EQIQDDCTGDED that corresponds to a fragment of apoB-100 (445-456). In this paper we studied how this peptide, as well as inhibitors and modulators of halogenating activity of MPO such as ceruloplasmin (CP), 4-aminobenzoic acid hydrazide (ABAH) and thiocyanate (SCN(-)) affect the accumulation of cholesterol and its esters in monocytes/macrophages after incubation with LDL subjected to different kinds of MPO-dependent oxidative/halogenating modification. In the presence of H2O2 and halides MPO causes stronger proatherogenic modification of LDL than exogenous reactive halogen species (HOCl and HOBr). Both monocytes, which differentiate into macrophages, and neutrophils secrete MPO in response to the presence of damaged LDL. The peptide EQIQDDCTGDED preventing interaction between MPO and LDL reduces the uptake of modified LDL and MPO by monocytes/macrophages and thus precludes the accumulation of intracellular cholesterol. Our results indicate that binding to MPO is important for LDL to become modified and acquire proatherogenic properties. The peptide EQIQDDCTGDED, CP, ABAH, and SCN(-) can play the role of anti-atherogenic factors reducing the deleterious effect of catalytically active MPO on LDL and accumulation of cholesterol in macrophages.
Collapse
Affiliation(s)
- Alexej V Sokolov
- Institute of Experimental Medicine of the N-W Branch of the Russian Academy of Medical Sciences, Saint-Petersburg, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia.
| | - Valeria A Kostevich
- Institute of Experimental Medicine of the N-W Branch of the Russian Academy of Medical Sciences, Saint-Petersburg, Russia; Research Institute of Physico-Chemical Medicine, Moscow, Russia
| | - Olga L Runova
- Institute of Experimental Medicine of the N-W Branch of the Russian Academy of Medical Sciences, Saint-Petersburg, Russia
| | | | - Vadim B Vasilyev
- Institute of Experimental Medicine of the N-W Branch of the Russian Academy of Medical Sciences, Saint-Petersburg, Russia; Saint-Petersburg State University, Saint-Petersburg, Russia
| | | | | |
Collapse
|
289
|
Nita C, Bala C, Porojan M, Hancu N. Fenofibrate improves endothelial function and plasma myeloperoxidase in patients with type 2 diabetes mellitus: an open-label interventional study. Diabetol Metab Syndr 2014; 6:30. [PMID: 24594096 PMCID: PMC3974011 DOI: 10.1186/1758-5996-6-30] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/13/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Fenofibrate offers a number of benefits on the cardiovascular system and it is plausible that its anti-inflammatory, anti-oxidant and anti-fibrotic effects and enhancement of cardiac metabolic performances may account for its direct cardioprotective effects.In this study we aimed to investigate the effect of fenofibrate on endothelial function assesed by vascular studies and levels of soluble E-selectin (sE-selectin) as well as the effect on plasma myeloperoxidase (MPO) in patients with type 2 diabetes mellitus (T2DM) without previous use of lipid-lowering medication. METHODS 27 patients (14 men and 13 women) with T2DM and good glycemic control (HbA1c: min 5.9%, max: 7.1%) treated with metformin monotherapy, without previous use of lipid-lowering medication were enrolled in this study. Vascular studies included measures of brachial artery diameter before and after release of a suprasystolic ischemia. FMD was calculated as the percent (%) change in arterial diameter following reactive hyperemia. Student's paired t test and Wilcoxon Signed Ranks Test were used to compare values before and after fenofibrate therapy. RESULTS Fenofibrate therapy significantly increased post ischemia mean brachial artery diameter at 60 s (from 4.7 [4.4; 5.0] mm to 4.9 [4.6; 5.2] mm, p = 0.01) and at 90 s (from 4.7 [4.4; 5.0] mm to 4.9 [4.6; 5.1], p = 0.02). FMD response to hyperaemia at 60 s increased with 4.5 ± 13.7% (median value pre- treatment: 22.2%, median value post- treatment 25.0%, z = -2.9, p = 0.004). After 8 weeks of fenofibrate therapy, plasma MPO levels decreased to 49.5 [30.3; 71.5] ng/ml (% change from baseline = 4.6%, z = -2.2, p = 0.03) and mean plasma sE-selectin levels decreased to 67.1 [54.4; 79.8] ng/ml, (% change from baseline = 2.6%, p = 0.03). CONCLUSION In patients with T2DM without previous treatment for dyslipidemia, short-term treatment with fenofibrate improved vascular endothelial function as demonstrated by increased post ischemia mean brachial artery diameter, increased FMD and decreased plasma sE-selectin and favorably affected plasma MPO levels. Therefore, fenofibrate may be considered a protective cardiovascular drug in this group of patients. TRIAL REGISTRATION (Australian New Zealand Clinical Trials Registry ANZCTR12612000734864).
Collapse
Affiliation(s)
- Cristina Nita
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Cornelia Bala
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Mihai Porojan
- Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 4-6 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Nicolae Hancu
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| |
Collapse
|
290
|
Molecular imaging of macrophage enzyme activity in cardiac inflammation. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014; 7:9258. [PMID: 24729833 DOI: 10.1007/s12410-014-9258-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular imaging is highly advantageous as various insidious inflammatory events can be imaged in a serial and quantitative fashion. Combined with the conventional imaging modalities like computed tomography (CT), magnetic resonance (MR) and nuclear imaging, it helps us resolve the extent of ongoing pathology, quantify inflammation and predict outcome. Macrophages are increasingly gaining importance as an imaging biomarker in inflammatory cardiovascular diseases. Macrophages, recruited to the site of injury, internalize necrotic or foreign material. Along with phagocytosis, activated macrophages release proteolytic enzymes like matrix metalloproteinases (MMPs) and cathepsins into the extracellular environment. Pro-inflammatory monocytes and macrophages also induce tissue oxidative damage through the inflammatory enzyme myeloperoxidase (MPO). In this review we will highlight recent advances in molecular macrophage imaging. Particular stress will be given to macrophage functional and enzymatic activity imaging which targets phagocytosis, proteolysis and myeloperoxidase activity imaging.
Collapse
|
291
|
Cardiac biomarkers in heart failure. Clin Biochem 2014; 47:327-37. [PMID: 24530339 DOI: 10.1016/j.clinbiochem.2014.01.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 01/25/2014] [Accepted: 01/27/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Heart failure is a syndrome characterized by the inability of the heart to meet the body's circulatory demands. Heart failure is a growing health issue worldwide and the prevalence of heart failure is expected to rise as populations age. Therapies and interventions for a variety of cardiac conditions continue to advance and biomarkers will play an increasing role in patient management. METHODS This is a review of the clinical research in blood based biomarkers for diagnosis, prognosis and therapeutic guidance of heart failure. The focus of this review is biomarkers that are currently available for clinical measurement, and their current and potential for applications for managing heart failure patients. RESULTS The various biologic pathways and physiologic processes of heart failure biomarkers represent a host of different including inflammation, remodeling, strain, neurohormonal activation, metabolism and cardiac myocyte injury. The clinical characteristics and applications of each heart failure biomarker are discussed. CONCLUSION As populations age and effective treatments and interventions for coronary artery disease improve, heart failure will increase in incidence and prevalence. Blood biomarkers will play an increasing role in the early diagnosis, therapeutic monitoring and management of heart failure patients in the future.
Collapse
|
292
|
Roed T, Kristoffersen US, Knudsen A, Wiinberg N, Lebech AM, Almdal T, Thomsen RW, Kjær A, Weis N. Increased prevalence of coronary artery disease risk markers in patients with chronic hepatitis C--a cross-sectional study. Vasc Health Risk Manag 2014; 10:55-62. [PMID: 24482574 PMCID: PMC3905100 DOI: 10.2147/vhrm.s53557] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective Chronic hepatitis C is a global health problem and has been associated with coronary artery disease. Our aim was to examine the prevalence of coronary artery disease risk markers including endothelial biomarkers in patients with chronic hepatitis C and matched comparisons without manifest cardiovascular disease or diabetes in a cross-sectional design. Methods Sixty patients with chronic hepatitis C (mean age 51 years) were recruited from the Department of Infectious Diseases at Copenhagen University Hospital, and compared with 60 age-matched non-hepatitis C virus-infected individuals from a general population survey. We examined traditional coronary artery disease risk factors, metabolic syndrome, carotid intima media thickness, and a range of endothelial biomarkers. Results Patients with chronic hepatitis C had more hypertension (40% versus 25%, prevalence ratio [PR] 1.6; 95% confidence interval [CI] 0.9–2.7) and smoked more (53% versus 38%, PR 1.4; 95% CI 0.9–2.1). The two groups had similar body mass index (mean 25.0 versus 25.7 kg/m2), whereas those with chronic hepatitis C had less dyslipidemia (including significantly lower low-density lipoprotein and cholesterol/high-density lipoprotein ratio), higher glycosylated hemoglobin level (mean 6.2 versus 5.7, difference of means 0.5; 95% CI 0.3–0.8), and a higher prevalence of metabolic syndrome (28% versus 18%, PR 1.6; 95% CI 0.8–3.0). Increased carotid intima media thickness above the standard 75th percentile was seen more frequently in chronic hepatitis C (9% versus 3%, PR 1.7; 95% CI 0.4–6.7), though difference of means was only 0.04 mm (95% CI 0.00–0.10). Patients with chronic hepatitis C had increased hsCRP (high-sensitivity C-reactive protein), sICAM-1 (soluble intercellular adhesion molecule-1), sVCAM-1 (soluble vascular cell adhesion molecule-1), and soluble E-selectin, but lower levels of tPAI-1 (tissue-type plasminogen activator inhibitor-1), MMP9 (matrix metallopeptidase 9), and MPO (myeloperoxidase) than their comparisons. Conclusion Our findings indicate that patients with chronic hepatitis C have increased prevalence of several coronary artery disease risk markers. These results may be important when evaluating the appropriateness of screening for coronary artery disease and its risk factors in chronic hepatitis C.
Collapse
Affiliation(s)
- Torsten Roed
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Ulrik Sloth Kristoffersen
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Knudsen
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark ; Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Wiinberg
- Department of Clinical Physiology, Frederiksberg Hospital, Copenhagen, Denmark
| | - Anne-Mette Lebech
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Reimar W Thomsen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark ; Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
293
|
|
294
|
Schuhmann CG, Hacker M, Jung P, Krötz F, Sohn HY. Myeloperoxidase is not useful for detecting stress inducible myocardial ischemia but may be indicative of the severity of coronary artery disease. Korean Circ J 2014; 44:10-5. [PMID: 24497884 PMCID: PMC3905110 DOI: 10.4070/kcj.2014.44.1.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/04/2013] [Accepted: 11/11/2013] [Indexed: 11/12/2022] Open
Abstract
Background and Objectives Elevated levels of myeloperoxidase (MPO) have been found in patients in different stages of coronary artery disease (CAD). The aim of this study was to assess whether the MPO liberation is increased by stress inducible myocardial ischemia and could be used to improve the diagnostic accuracy of non-invasive evaluation for myocardial ischemia. Subjects and Methods Seventy-six patients with suspected myocardial ischemia who underwent stress myocardial perfusion scintigraphy (MPS) were enrolled. 59 patients with an acute coronary syndrome (ACS) who received a percutaneous coronary intervention along with 12 healthy volunteers were also included in the study. In every subject the MPO plasma levels were assessed by enzyme linked immunosorbent assay. In patients undergoing MPS, the MPO levels were measured serially before and after the stress testing. Results Of the 76 patients undergoing MPS, 38 were diagnosed with a stress inducible myocardial ischemia. The patients with a stress induced ischemia had significantly higher basal MPO levels than those without it (32±3 ng/mL vs. 24±4 ng/mL, p=0.03). However, there was no relevant change in the MPO levels after the stress test compared to the baseline. The patients with ACS showed significantly higher MPO levels than the patients undergoing MPS (131±14 ng/mL vs. 28±2 ng/mL, p<0.01) and the healthy subjects (131±14 ng/mL vs. 26±2 ng/mL, p<0.01). Conclusion Since the MPO plasma levels did not increase after the stress MPS, MPO appears not to be a useful biomarker for detecting a stress inducible myocardial ischemia. Yet, the MPO levels correlate with the different stages of CAD and may hold significance as an indicator for its clinical severity.
Collapse
Affiliation(s)
| | - Marcus Hacker
- Department of Nuclear Medicine, University Hospital of the Ludwig Maximilian University of Munich, Munich, Germany
| | - Philip Jung
- Department of Cardiology, Medizinische Klinik I, Munich, Germany
| | - Florian Krötz
- Department of Cardiology, Medizinische Klinik I, Munich, Germany
| | - Hae-Young Sohn
- Department of Cardiology, Medizinische Klinik I, Munich, Germany
| |
Collapse
|
295
|
Niccoli G, Montone RA, Cataneo L, Cosentino N, Gramegna M, Refaat H, Porto I, Burzotta F, Trani C, Leone AM, Severino A, Crea F. Morphological-biohumoral correlations in acute coronary syndromes: pathogenetic implications. Int J Cardiol 2014; 171:463-6. [PMID: 24439867 DOI: 10.1016/j.ijcard.2013.12.238] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/30/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Giampaolo Niccoli
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy.
| | - Rocco A Montone
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Leonardo Cataneo
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Nicola Cosentino
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Mario Gramegna
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Hesham Refaat
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Italo Porto
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Burzotta
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Carlo Trani
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonio M Leone
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Severino
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Crea
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
296
|
Toth PP, McCullough PA, Wegner MS, Colley KJ. Lipoprotein-associated phospholipase A2: role in atherosclerosis and utility as a cardiovascular biomarker. Expert Rev Cardiovasc Ther 2014; 8:425-38. [DOI: 10.1586/erc.10.18] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
297
|
Myeloperoxidase upregulates endothelin receptor type B expression. J Mol Cell Cardiol 2014; 69:76-82. [PMID: 24417960 DOI: 10.1016/j.yjmcc.2013.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 11/29/2013] [Accepted: 12/10/2013] [Indexed: 11/23/2022]
Abstract
Neutrophil recruitment and activation are principal events in inflammation. Upon activation neutrophils release myeloperoxidase (MPO), a heme enzyme, which binds to and transcytoses endothelial cells. Whereas the significance of the subendothelial deposition of MPO has evolved as a critical prerequisite for the enzyme's suppression of nitric oxide (NO⋅) bioavailability, the functional consequences of MPO binding to and interaction with endothelial and smooth muscle cells remain poorly understood. Cultured human endothelial cells (HUVECs) were exposed to MPO. Gene expression of the endothelin receptor type B (ETRB), which is critically involved not only in endothelin-1 clearance, but also in endothelin-mediated vasoconstriction, was significantly increased. Real time PCR, Western blotting and immunofluorescence confirmed up-regulation of ETRB in MPO-treated endothelial cells. Inhibition of MPO's enzymatic activity blunted the increase in ETRB protein expression. Treatment of the cells with the MAP kinase inhibitors PD98059 or SB203580 indicates that MPO activates ETRB expression via MAP kinase pathways. On human smooth muscle cells (HAoSMCs), which not only express the endothelin receptor type B (ETRB) but also express the endothelin receptor type A (ETRA), MPO also stimulated ETRB expression as opposed to ETRA expression, which remained unchanged. Functional ex vivo organ bath chamber studies with MPO-incubated rat femoral artery sections revealed increased ETRB agonist dependent constriction. Binding of MPO to endothelial and vascular smooth muscle cells increases expression of the endothelin receptor type B (ETRB) via classical MAP kinase pathways. This suggests that MPO not only affects vasomotion by reducing the bioavailability of vasodilating molecules but also by increasing responsiveness to vasoconstrictors, further advocating for MPO as a central, leukocyte-derived regulator of vascular tone.
Collapse
|
298
|
Begenik H, Soyoral YU, Erkoc R, Emre H, Taskın A, Tasdemir M, Aslan M. Serum malondialdehyde levels, myeloperoxidase and catalase activities in patients with nephrotic syndrome. Redox Rep 2014; 18:107-12. [PMID: 23746122 DOI: 10.1179/1351000213y.0000000048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVES Some studies have indicated the pathophysiological importance of reactive oxygen species (ROS) in patients with nephrotic syndrome. Myeloperoxidase (MPO) is a leukocyte-derived enzyme-generating ROS that has been proposed to exert a wide array of pro-atherogenic effects throughout all stages of the atherosclerotic process. The aim of this study was to investigate the serum malondialdehyde (MDA) levels, MPO and catalase activities in patients with adult nephrotic syndrome. PATIENTS AND METHOD s Twenty-four patients with nephrotic syndrome and 24 healthy controls were enrolled. Serum MPO activity, catalase activity, and MDA levels were assessed. RESULTS Serum MPO activity and MDA levels were significantly higher in patients with nephrotic syndrome than controls (both, P<0.001), while catalase activity was significantly lower (P<0.001). Serum catalase activity was found to be significantly correlated with MPO activity (r=-0.417, P=0.003) and MDA levels (r=-0.532, P=0.007). The serum MDA levels were also found to be significantly correlated with MPO activity (r=0.419, P=0.003). CONCLUSIONS We concluded that serum MPO activity and oxidative stress were increased and that serum catalase activity was decreased in patients with adult nephrotic syndrome. In addition, these results indicate that increased MPO activity is associated with an oxidant-antioxidant imbalance that may contribute to atherosclerosis in patients with adult nephrotic syndrome.
Collapse
Affiliation(s)
- Huseyin Begenik
- Department Nephrology, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | | | | | | | | | | | | |
Collapse
|
299
|
Laborde CM, Mourino-Alvarez L, Posada-Ayala M, Alvarez-Llamas G, Serranillos-Reus MG, Moreu J, Vivanco F, Padial LR, Barderas MG. Plasma metabolomics reveals a potential panel of biomarkers for early diagnosis in acute coronary syndrome. Metabolomics 2014; 10:414-424. [PMID: 25814918 PMCID: PMC4363481 DOI: 10.1007/s11306-013-0595-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 10/15/2013] [Indexed: 12/22/2022]
Abstract
Discovery of new biomarkers is critical for early diagnosis of acute coronary syndrome (ACS). Recent advances in metabolomic technologies have drastically enhanced the possibility of improving the knowledge of its physiopathology through the identification of the altered metabolic pathways. In this study, analyses of peripheral plasma from non-ST segment elevation ACS patients and healthy controls by gas chromatography-mass spectrometry (GC-MC) permitted the identification of 15 metabolites with statistical differences (p < 0.05) between experimental groups. Additionally, validation by GC-MC and liquid chromatography-MC permitted us to identify a potential panel of biomarkers formed by 5-OH-tryptophan, 2-OH-butyric acid and 3-OH-butyric acid. This panel of biomarkers reflects the oxidative stress and the hypoxic state that suffers the myocardial cells and consequently constitutes a metabolomic signature of the atherogenesis process that could be used for early diagnosis of ACS.
Collapse
Affiliation(s)
- Carlos M. Laborde
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | | | | | | | - José Moreu
- Department of Hemodynamic, Hospital Virgen de la Salud, SESCAM, Toledo, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Department of Biochemistry and Molecular Biology I, Facultad de Biología, UCM, Madrid, Spain
| | - Luis R. Padial
- Department of Cardiology, Hospital Virgen de la Salud, SESCAM, Toledo, Spain
| | - María G. Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
- Proteomic Unit, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| |
Collapse
|
300
|
Kormi I, Alfakry H, Tervahartiala T, Pussinen PJ, Sinisalo J, Sorsa T. The effect of prolonged systemic doxycycline therapy on serum tissue degrading proteinases in coronary bypass patients: a randomized, double-masked, placebo-controlled clinical trial. Inflamm Res 2013; 63:329-34. [PMID: 24378958 DOI: 10.1007/s00011-013-0704-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/10/2013] [Accepted: 11/25/2013] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Serum matrix metalloproteinases (MMP-8, MMP-7) and their regulators may be associated with the risk of incident cardiovascular disease events. Doxycycline can be used as matrix metalloproteinase (MMP) inhibitor independent of its antimicrobial activity. We aimed to investigate serum inflammatory biomarkers during 4 months of doxycycline therapy in coronary bypass patients. MATERIALS AND METHODS Thirty-one non-smoking men who had previous coronary bypass surgery were randomly assigned to receive placebo or 100 mg doxycycline daily for 4 months. Serum samples were collected at baseline before the treatment, and at 2, 4, and 10 months. Serum levels of MMP-7, tissue inhibitor of matrix metalloproteinase (TIMP)-1, myeloperoxidase, and neutrophil elastase were analyzed with enzyme-linked immunosorbent assay, MMP-8 by immunofluorometric assay, and C-reactive protein by rate nephelometry. RESULTS At baseline, no significant differences existed between the two groups. Serum levels of MMP-8, MMP-7, and MMP-8/TIMP-1 were and remained lower (p = 0.034, p = 0.041, and NS) in the doxycycline group relative to the placebo group at 4 months of follow-up. CONCLUSIONS Doxycycline decreases the systemic inflammatory burden in patients with myocardial infarction and especially down-regulates MMP-7, MMP-8, and MMP-8/TIMP-1. Doxycycline might prevent or reduce the risk of secondary myocardial infarctions by providing a systemic anti-proteolytic and -inflammatory shield.
Collapse
Affiliation(s)
- Immi Kormi
- Oral and Maxillofacial Department, Oulu University Hospital, Oulu, Finland
| | | | | | | | | | | |
Collapse
|