1
|
Geissen S, Braumann S, Adler J, Nettersheim FS, Mehrkens D, Hof A, Guthoff H, von Stein P, Witkowski S, Gerdes N, Tellkamp F, Krüger M, Isermann L, Trifunovic A, Bunck AC, Mollenhauer M, Winkels H, Adam M, Klinke A, Buch G, Ten Cate V, Hellmich M, Kelm M, Rudolph V, Wild PS, Rosenkranz S, Baldus S. Inhibition of myeloperoxidase to treat left ventricular dysfunction in non-ischaemic cardiomyopathy. Eur J Heart Fail 2024; 26:2269-2281. [PMID: 39212229 DOI: 10.1002/ejhf.3435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
AIMS Non-ischaemic cardiomyopathy (NICMP), an incurable disease terminating in systolic heart failure (heart failure with reduced ejection fraction [HFrEF]), causes immune activation, however anti-inflammatory treatment strategies so far have failed to alter the course of this disease. Myeloperoxidase (MPO), the principal enzyme in neutrophils, has cytotoxic, pro-fibrotic and nitric oxide oxidizing effects. Whether MPO inhibition ameliorates the phenotype in NICMP remains elusive. METHODS AND RESULTS Prognostic information from MPO was derived from proteomic data of a large human cardiovascular health cohort (n = 3289). In a murine model of NICMP, we studied the mechanisms of MPO in this disease. In a case series, the MPO inhibitor was also evaluated in NICMP patients. Individuals with increased MPO revealed higher long-term mortality and worsening of heart failure, with impaired prognosis when MPO increased during follow-up. MPO infusion attenuated left ventricular ejection fraction (LVEF) in mice with NICMP, whereas genetic ablation or inhibition of MPO decreased systemic vascular resistance (SVR, 9.4 ± 0.7 mmHg*min/ml in NICMP vs. 6.7 ± 0.8 mmHg*min/ml in NICMP/Mpo-/-mice, n = 8, p = 0.006, data expressed as mean ± standard error of the mean) and improved left ventricular function (LVEF 30.3 ± 2.2% in NICMP vs. 40.7 ± 1.1% in NICMP/Mpo-/- mice, n = 16, p < 0.0001). Four patients diagnosed with NICMP and treated with an MPO inhibitor over 12 weeks showed increase in LVEF, decline in natriuretic peptides and improved 6-min walking distance. MPO inhibitor-related changes in the proteome of NICMP patients predicted reduced mortality when related to the changes in the proteome of the above referenced cardiovascular health cohort. CONCLUSIONS Myeloperoxidase predicts long-term outcome in HFrEF and its inhibition elicits systemic anti-inflammatory and vasodilating effects which translate into improved left ventricular function. MPO inhibition deserves further evaluation as a novel, complementary treatment strategy for HFrEF.
Collapse
Affiliation(s)
- Simon Geissen
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Simon Braumann
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Joana Adler
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Felix Sebastian Nettersheim
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Dennis Mehrkens
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Alexander Hof
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Henning Guthoff
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Philipp von Stein
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
| | - Sven Witkowski
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Frederik Tellkamp
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, Germany
- University of Cologne, Department of Biology, Institute for Genetics, Cologne, Germany
| | - Marcus Krüger
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, Germany
- University of Cologne, Department of Biology, Institute for Genetics, Cologne, Germany
| | - Lea Isermann
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, Germany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexander C Bunck
- Department of Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Martin Mollenhauer
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Matti Adam
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Anna Klinke
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Gregor Buch
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Vincent Ten Cate
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Martin Hellmich
- Institute of Medical Statistics and Computational Biology (IMSB), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Volker Rudolph
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stephan Rosenkranz
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Stephan Baldus
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| |
Collapse
|
2
|
Singh KK, Gupta A, Forstner D, Guettler J, Ahrens MS, Prakasan Sheeja A, Fatima S, Shamkeeva S, Lia M, Dathan-Stumpf A, Hoffmann N, Shahzad K, Stepan H, Gauster M, Isermann B, Kohli S. LMWH prevents thromboinflammation in the placenta via HBEGF-AKT signaling. Blood Adv 2024; 8:4756-4766. [PMID: 38941535 PMCID: PMC11457404 DOI: 10.1182/bloodadvances.2023011895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/24/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT Low molecular weight heparins (LMWH) are used to prevent or treat thromboembolic events during pregnancy. Although studies suggest an overall protective effect of LMWH in preeclampsia (PE), their use in PE remains controversial. LMWH may convey beneficial effects in PE independent of their anticoagulant activity, possibly by inhibiting inflammation. Here, we evaluated whether LMWH inhibit placental thromboinflammation and trophoblast NLRP3 inflammasome activation. Using an established procoagulant extracellular vesicle-induced and platelet-dependent PE-like mouse model, we show that LMWH reduces pregnancy loss and trophoblast inflammasome activation, restores altered trophoblast differentiation, and improves trophoblast proliferation in vivo and in vitro. Moreover, LMWH inhibits platelet-independent trophoblast NLRP3 (NLR family pyrin domain containing 3) inflammasome activation. Mechanistically, LMWH activates via heparin-binding epidermal growth factor (HBEGF) signaling the PI3-kinase-AKT pathway in trophoblasts, thus preventing inflammasome activation. In human PE placental explants, inflammasome activation and PI3-kinase-AKT signaling events were reduced with LMWH treatment compared with those without LMWH treatment. Thus, LMWH inhibits sterile inflammation via the HBEGF signaling pathway in trophoblasts and ameliorates PE-associated complications. These findings suggest that drugs targeting the inflammasome may be evaluated in PE and identify a signaling mechanism through which LMWH ameliorates PE, thus providing a rationale for the use of LMWH in PE.
Collapse
Affiliation(s)
- Kunal Kumar Singh
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Mirjam Susanne Ahrens
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Akshay Prakasan Sheeja
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Saikal Shamkeeva
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Massimiliano Lia
- Department of Obstetrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Anne Dathan-Stumpf
- Department of Obstetrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Nikola Hoffmann
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Holger Stepan
- Department of Obstetrics, University of Leipzig Medical Center, Leipzig, Germany
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| |
Collapse
|
3
|
Quinn M, Zhang RYK, Bello I, Rye KA, Thomas SR. Myeloperoxidase as a Promising Therapeutic Target after Myocardial Infarction. Antioxidants (Basel) 2024; 13:788. [PMID: 39061857 PMCID: PMC11274265 DOI: 10.3390/antiox13070788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) remain leading causes of death and disability worldwide. CAD begins with the formation of atherosclerotic plaques within the intimal layer of the coronary arteries, a process driven by persistent arterial inflammation and oxidation. Myeloperoxidase (MPO), a mammalian haem peroxidase enzyme primarily expressed within neutrophils and monocytes, has been increasingly recognised as a key pro-inflammatory and oxidative enzyme promoting the development of vulnerable coronary atherosclerotic plaques that are prone to rupture, and can precipitate a MI. Mounting evidence also implicates a pathogenic role for MPO in the inflammatory process that follows a MI, which is characterised by the rapid infiltration of activated neutrophils into the damaged myocardium and the release of MPO. Excessive and persistent cardiac inflammation impairs normal cardiac healing post-MI, resulting in adverse cardiac outcomes and poorer long-term cardiac function, and eventually heart failure. This review summarises the evidence for MPO as a significant oxidative enzyme contributing to the inappropriate inflammatory responses driving the progression of CAD and poor cardiac healing after a MI. It also details the proposed mechanisms underlying MPO's pathogenic actions and explores MPO as a novel therapeutic target for the treatment of unstable CAD and cardiac damage post-MI.
Collapse
Affiliation(s)
| | | | | | | | - Shane R. Thomas
- Cardiometabolic Disease Research Group, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
4
|
Yiu JYT, Hally KE, Larsen PD, Holley AS. Neutrophil-Enriched Biomarkers and Long-Term Prognosis in Acute Coronary Syndrome: a Systematic Review and Meta-analysis. J Cardiovasc Transl Res 2024; 17:426-447. [PMID: 37594719 PMCID: PMC11052791 DOI: 10.1007/s12265-023-10425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
Activated neutrophils release a range of inflammatory products that represent potential biomarkers, and there is interest in the prognostic value of these in acute coronary syndrome (ACS) patients. We conducted a systematic review to examine neutrophil-enriched biomarkers and the occurrence of major adverse cardiovascular events (MACE) in patients with ACS. We identified twenty-seven studies including 17,831 patients with ACS. The most studied biomarkers were neutrophil gelatinase-associated lipocalin (NGAL) and myeloperoxidase (MPO). Meta-analyses showed that elevated NGAL was associated with higher MACE rates (unadjusted risk ratio (RR) 1.52, 95% CI 1.12-2.06, p = 0.006) as were elevated MPO levels (unadjusted RR 1.61, 95% CI 1.22-2.13, p = 0.01). There was limited data suggesting that increased levels of calprotectin, proteinase-3 and double-stranded DNA were also associated with MACE. These results suggest that higher levels of neutrophil-enriched biomarkers may be predictive of MACE in patients with ACS, although higher-quality studies are needed to confirm these observations.
Collapse
Affiliation(s)
- Jaquelina Y T Yiu
- Wellington Cardiovascular Research Group, Department of Surgery & Anaesthesia, University of Otago, PO Box 7343, Wellington, New Zealand
| | - Kathryn E Hally
- Wellington Cardiovascular Research Group, Department of Surgery & Anaesthesia, University of Otago, PO Box 7343, Wellington, New Zealand
| | - Peter D Larsen
- Wellington Cardiovascular Research Group, Department of Surgery & Anaesthesia, University of Otago, PO Box 7343, Wellington, New Zealand
| | - Ana S Holley
- Wellington Cardiovascular Research Group, Department of Surgery & Anaesthesia, University of Otago, PO Box 7343, Wellington, New Zealand.
| |
Collapse
|
5
|
Halldorsdottir H, Lindbom L, Ebberyd A, Oldner A, Weitzberg E. The effect of heparins on plasma concentration of heparin-binding protein: a pilot study. BJA OPEN 2024; 9:100256. [PMID: 38318270 PMCID: PMC10839136 DOI: 10.1016/j.bjao.2023.100256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/17/2023] [Indexed: 02/07/2024]
Abstract
Background Neutrophil-derived heparin-binding protein (HBP) plays a role in the pathophysiology of impaired endothelial dysfunction during inflammation. HBP has been suggested as a predictor of organ dysfunction and disease progression in sepsis. We investigated the effects of heparins on plasma concentrations of HBP in patients undergoing surgery. Methods We studied three groups of patients receiving heparins during or after surgery. The vascular surgery group received 3000-7500 U, whereas the cardiac surgery group received 27 500-40 000 U. After major general surgery, the third group received 5000 U of low-molecular-weight heparin (LMWH) subcutaneously. Serial plasma HBP concentrations were measured after these treatments with two different methods: Axis-Shield ELISA and Joinstar FIC-Q100. In addition, plasma myeloperoxidase and syndecan-1 were measured in the cardiac surgery group. Results During vascular surgery, heparin induced a six-fold increase in HBP within 2 min, from 3.6 (2.4-5.4) to 21.4 (9.0-35.4) ng ml-1 (P<0.001). During cardiac surgery, the higher dose of heparin elevated HBP concentrations from 5.3 (2.7-6.1) to 48.7 (38.4-70.1) ng ml-1 (P<0.0001) within 3 min. Patients receiving LMWH showed an increase from a baseline of 5.7 (3.7-12.1) ng ml-1 to a peak HBP concentration of 14.8 (9.5-18.1) ng ml-1 (P<0.0001) after 3 h. Plasma concentrations of myeloperoxidase, but not syndecan-1, also responded with a rapid increase after heparin. There was a strong correlation between the two methods for HBP analysis (r=0.94). Conclusions Plasma concentrations of HBP increased rapidly and dose-dependently after heparin administration. Subcutaneous administration of LMWH increases plasma HBP, but to a lesser degree. Clinical trial registration ClinicalTrials.gov identifier: NCT04146493.
Collapse
Affiliation(s)
- Halla Halldorsdottir
- Department of Clinical Sciences, Division of Anaesthesia and Intensive Care, Karolinska Institutet, Danderyd Hospital, Danderyd, Sweden
- Department of Physiology and Pharmacology, Section of Anaesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Lindbom
- Department of Physiology and Pharmacology, Section of Anaesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Anette Ebberyd
- Department of Physiology and Pharmacology, Section of Anaesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Anders Oldner
- Department of Physiology and Pharmacology, Section of Anaesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Section of Anaesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Adam LN, Al-Habib OAM, Oraha AY, Shekha MS. Genetic and clinical study of myeloperoxidase's association with coronary artery disease. Egypt Heart J 2024; 76:27. [PMID: 38383869 PMCID: PMC10881921 DOI: 10.1186/s43044-024-00457-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/16/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Unraveling myeloperoxidase's (MPO) correlation with coronary artery disease (CAD) and genetic variations, this study seeks to enhance diagnostic precision and therapeutic strategies. RESULTS CAD patients were found to be older and more male than controls. Several clinical parameters, including glucose, total bilirubin, alkaline phosphatase, creatinine, and troponin levels, showed significant variations. Moreover, CAD patients had lower red cell distribution width (RDW%) and mean platelet volume (MPV) than controls. Serum MPO levels did not differ significantly between CAD patients and controls, and no correlation was found with other clinical parameters except for glucose, creatinine, and total bilirubin. CONCLUSIONS The data suggest that serum MPO levels are not substantially related to CAD patients, as indicated by lower MPO levels in CAD patients compared to controls. While highlighting the potential of MPV and RDW% as predictors of severe atherosclerosis in CAD. Further research is needed to validate the diagnostic and prognostic value of RDW%, MPV, and MPO levels in CAD. TRIAL REGISTRATION 15092021-9-12. Registered 15 September 2021.
Collapse
Affiliation(s)
- Lina N Adam
- Department of Biology, College of Science, University of Zakho, Duhok, Kurdistan Region, Iraq.
| | - Omar A M Al-Habib
- Department of Biology, College of Science, University of Nawroz, Duhok, Kurdistan Region, Iraq
| | - Ashur Y Oraha
- Department of Cardiothoracic and Vascular Surgery, College of Medicine, University of Duhok, Duhok, Kurdistan Region, Iraq
| | - Mudhir S Shekha
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
7
|
Gimblet CJ, Ernst JW, Bos KD, Stroud AK, Donato AJ, Jalal DI, Pierce GL. Effect of acute heparin administration on glycocalyx thickness and endothelial function in healthy younger adults. J Appl Physiol (1985) 2024; 136:330-336. [PMID: 38126088 PMCID: PMC11212829 DOI: 10.1152/japplphysiol.00767.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023] Open
Abstract
The endothelial glycocalyx is a dynamic, gel-like layer that is critical to normal vascular endothelial function. Heparin impairs the endothelial glycocalyx and reduces vascular endothelial function in a murine model; however, this has yet to be tested in healthy humans. We hypothesized that a single bolus dose of heparin would increase circulating glycocalyx components and decrease endothelial glycocalyx thickness resulting in blunted brachial artery vasodilation in healthy younger adults. Healthy adults (n = 19, aged 18-39 yr, 53% female) underwent measurements of the endothelial glycocalyx and vascular endothelial function at baseline and after a single bolus 5,000 U dose of heparin. The glycocalyx components syndecan-1 and heparan sulfate were measured from plasma samples using enzyme-linked immunosorbent assays. Glycocalyx thickness was determined as perfused boundary region (PBR) in sublingual microvessels using the GlycoCheck. Endothelial function was measured via ultrasonography and quantified as brachial artery flow-mediated dilation (FMD). Following acute heparin administration, there was no increase in syndecan-1 or heparan sulfate (P = 0.90 and P = 0.49, respectively). In addition, there was no change in PBR 4-7 µm (P = 0.55), PBR 10-25 µm (P = 0.63), or 4-25 µm (P = 0.49) after heparin treatment. Furthermore, we did not observe a change in FMDmm (P = 0.23), FMD% (P = 0.35), or plasma nitrite concentrations (P = 0.10) in response to heparin. Finally, time to peak dilation and peak FMD normalized to shear stress were unchanged following heparin (P = 0.59 and P = 0.21, respectively). Our pilot study suggests that a single bolus intravenous dose of heparin does not result in endothelial glycocalyx degradation or vascular endothelial dysfunction in healthy younger adults.NEW & NOTEWORTHY The endothelial glycocalyx's role in modulating vascular endothelial dysfunction with aging and disease is becoming increasingly recognized. This study presents novel findings that acute heparin administration is not a feasible method to experimentally degrade the endothelial glycocalyx and measure concurrent changes in vascular endothelial function in healthy humans. Alternative approaches will be needed to translate findings from preclinical studies and test the effects of acute endothelial glycocalyx degradation on vascular endothelial function in humans.
Collapse
Affiliation(s)
- Colin J Gimblet
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
| | - Jackson W Ernst
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
| | - Kyle D Bos
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
| | - Amy K Stroud
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Diana I Jalal
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
- Center for Access and Delivery Research and Evaluation, Iowa City VA Medical Center, Iowa City, Iowa, United States
| | - Gary L Pierce
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
8
|
Cosic-Mujkanovic N, Valadez-Cosmes P, Maitz K, Lueger A, Mihalic ZN, Runtsch MC, Kienzl M, Davies MJ, Chuang CY, Heinemann A, Schicho R, Marsche G, Kargl J. Myeloperoxidase Alters Lung Cancer Cell Function to Benefit Their Survival. Antioxidants (Basel) 2023; 12:1587. [PMID: 37627581 PMCID: PMC10451743 DOI: 10.3390/antiox12081587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Myeloperoxidase (MPO) is a neutrophil-derived enzyme that has been recently associated with tumour development. However, the mechanisms by which this enzyme exerts its functions remain unclear. In this study, we investigated whether myeloperoxidase can alter the function of A549 human lung cancer cells. We observed that MPO promoted the proliferation of cancer cells and inhibited their apoptosis. Additionally, it increased the phosphorylation of AKT and ERK. MPO was rapidly bound to and internalized by A549 cells, retaining its enzymatic activity. Furthermore, MPO partially translocated into the nucleus and was detected in the chromatin-enriched fraction. Effects of MPO on cancer cell function could be reduced when MPO uptake was blocked with heparin or upon inhibition of the enzymatic activity with the MPO inhibitor 4-aminobenzoic acid hydrazide (4-ABAH). Lastly, we have shown that tumour-bearing mice treated with 4-ABAH had reduced tumour burden when compared to control mice. Our results highlight the role of MPO as a neutrophil-derived enzyme that can alter the function of lung cancer cells.
Collapse
Affiliation(s)
- Nejra Cosic-Mujkanovic
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Paulina Valadez-Cosmes
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Kathrin Maitz
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Anna Lueger
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Zala N. Mihalic
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Marah C. Runtsch
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Melanie Kienzl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Michael J. Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Christine Y. Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Rudolf Schicho
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
9
|
Inghardt T, Antonsson T, Ericsson C, Hovdal D, Johannesson P, Johansson C, Jurva U, Kajanus J, Kull B, Michaëlsson E, Pettersen A, Sjögren T, Sörensen H, Westerlund K, Lindstedt EL. Discovery of AZD4831, a Mechanism-Based Irreversible Inhibitor of Myeloperoxidase, As a Potential Treatment for Heart Failure with Preserved Ejection Fraction. J Med Chem 2022; 65:11485-11496. [PMID: 36005476 PMCID: PMC9469207 DOI: 10.1021/acs.jmedchem.1c02141] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Myeloperoxidase is a promising therapeutic target for treatment of patients suffering from heart failure with preserved ejection fraction (HFpEF). We aimed to discover a covalent myeloperoxidase inhibitor with high selectivity for myeloperoxidase over thyroid peroxidase, limited penetration of the blood-brain barrier, and pharmacokinetics suitable for once-daily oral administration at low dose. Structure-activity relationship, biophysical, and structural studies led to prioritization of four compounds for in-depth safety and pharmacokinetic studies in animal models. One compound (AZD4831) progressed to clinical studies on grounds of high potency (IC50, 1.5 nM in vitro) and selectivity (>450-fold vs thyroid peroxidase in vitro), the mechanism of irreversible inhibition, and the safety profile. Following phase 1 studies in healthy volunteers and a phase 2a study in patients with HFpEF, a phase 2b/3 efficacy study of AZD4831 in patients with HFpEF started in 2021.
Collapse
Affiliation(s)
- Tord Inghardt
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Thomas Antonsson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Cecilia Ericsson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Daniel Hovdal
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Petra Johannesson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Carina Johansson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Ulrik Jurva
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Johan Kajanus
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Bengt Kull
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Erik Michaëlsson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Anna Pettersen
- Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Tove Sjögren
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Henrik Sörensen
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Kristina Westerlund
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 50 Mölndal, Gothenburg, Sweden
| |
Collapse
|
10
|
Valadez-Cosmes P, Raftopoulou S, Mihalic ZN, Marsche G, Kargl J. Myeloperoxidase: Growing importance in cancer pathogenesis and potential drug target. Pharmacol Ther 2021; 236:108052. [PMID: 34890688 DOI: 10.1016/j.pharmthera.2021.108052] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
Myeloperoxidase is a heme-peroxidase which makes up approximately 5% of the total dry cell weight of neutrophils where it is predominantly found in the primary (azurophilic) granules. Other cell types, such as monocytes and certain macrophage subpopulations also contain myeloperoxidase, but to a much lesser extent. Initially, the function of myeloperoxidase had been mainly associated with its ability as a catalyzer of reactive oxidants that help to clear pathogens. However, over the past years non-canonical functions of myeloperoxidase have been described both in health and disease. Attention has been specially focused on inflammatory diseases, in which an exacerbate infiltration of leukocytes can favor a poorly-controlled production and release of myeloperoxidase and its oxidants. There is compelling evidence that myeloperoxidase derived oxidants contribute to tissue damage and the development and propagation of acute and chronic vascular inflammation. Recently, neutrophils have attracted much attention within the large diversity of innate immune cells that are part of the tumor microenvironment. In particular, neutrophil-derived myeloperoxidase may play an important role in cancer development and progression. This review article aims to provide a comprehensive overview of the roles of myeloperoxidase in the development and progression of cancer. We propose future research approaches and explore prospects of inhibiting myeloperoxidase as a strategy to fight against cancer.
Collapse
Affiliation(s)
- Paulina Valadez-Cosmes
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Sofia Raftopoulou
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Zala Nikita Mihalic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
11
|
Hawkins CL, Davies MJ. Role of myeloperoxidase and oxidant formation in the extracellular environment in inflammation-induced tissue damage. Free Radic Biol Med 2021; 172:633-651. [PMID: 34246778 DOI: 10.1016/j.freeradbiomed.2021.07.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
The heme peroxidase family generates a battery of oxidants both for synthetic purposes, and in the innate immune defence against pathogens. Myeloperoxidase (MPO) is the most promiscuous family member, generating powerful oxidizing species including hypochlorous acid (HOCl). Whilst HOCl formation is important in pathogen removal, this species is also implicated in host tissue damage and multiple inflammatory diseases. Significant oxidant formation and damage occurs extracellularly as a result of MPO release via phagolysosomal leakage, cell lysis, extracellular trap formation, and inappropriate trafficking. MPO binds strongly to extracellular biomolecules including polyanionic glycosaminoglycans, proteoglycans, proteins, and DNA. This localizes MPO and subsequent damage, at least partly, to specific sites and species, including extracellular matrix (ECM) components and plasma proteins/lipoproteins. Biopolymer-bound MPO retains, or has enhanced, catalytic activity, though evidence is also available for non-catalytic effects. These interactions, particularly at cell surfaces and with the ECM/glycocalyx induce cellular dysfunction and altered gene expression. MPO binds with higher affinity to some damaged ECM components, rationalizing its accumulation at sites of inflammation. MPO-damaged biomolecules and fragments act as chemo-attractants and cell activators, and can modulate gene and protein expression in naïve cells, consistent with an increasing cycle of MPO adhesion, activity, damage, and altered cell function at sites of leukocyte infiltration and activation, with subsequent tissue damage and dysfunction. MPO levels are used clinically both diagnostically and prognostically, and there is increasing interest in strategies to prevent MPO-mediated damage; therapeutic aspects are not discussed as these have been reviewed elsewhere.
Collapse
Affiliation(s)
- Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
12
|
Cullivan S, Murphy CA, Weiss L, Comer SP, Kevane B, McCullagh B, Maguire PB, Ní Ainle F, Gaine SP. Platelets, extracellular vesicles and coagulation in pulmonary arterial hypertension. Pulm Circ 2021; 11:20458940211021036. [PMID: 34158919 PMCID: PMC8182202 DOI: 10.1177/20458940211021036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Pulmonary arterial hypertension is a rare disease of the pulmonary vasculature, characterised pathologically by proliferation, remodelling and thrombosis in situ. Unfortunately, existing therapeutic interventions do not reverse these findings and the disease continues to result in significant morbidity and premature mortality. A number of haematological derangements have been described in pulmonary arterial hypertension which may provide insights into the pathobiology of the disease and opportunities to explore new therapeutic pathways. These include quantitative and qualitative platelet abnormalities, such as thrombocytopaenia, increased mean platelet volume and altered platelet bioenergetics. Furthermore, a hypercoagulable state and aberrant negative regulatory pathways can be observed, which could contribute to thrombosis in situ in distal pulmonary arteries and arterioles. Finally, there is increasing interest in the role of extracellular vesicle autocrine and paracrine signalling in pulmonary arterial hypertension, and their potential utility as biomarkers and novel therapeutic targets. This review focuses on the potential role of platelets, extracellular vesicles and coagulation pathways in the pathobiology of pulmonary arterial hypertension. We highlight important unanswered clinical questions and the implications of these observations for future research and pulmonary arterial hypertension-directed therapies.
Collapse
Affiliation(s)
- Sarah Cullivan
- National Pulmonary Hypertension Unit, Mater
Misericordiae University Hospital, Dublin, Ireland
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
| | - Claire A. Murphy
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
- Department of Neonatology, Rotunda Hospital, Dublin,
Ireland
| | - Luisa Weiss
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
| | - Shane P. Comer
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
| | - Barry Kevane
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae
University Hospital, Dublin, Ireland
| | - Brian McCullagh
- National Pulmonary Hypertension Unit, Mater
Misericordiae University Hospital, Dublin, Ireland
| | - Patricia B. Maguire
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
| | - Fionnuala Ní Ainle
- Conway-SPHERE Research Group, Conway Institute,
University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae
University Hospital, Dublin, Ireland
| | - Sean P. Gaine
- National Pulmonary Hypertension Unit, Mater
Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
13
|
Maiocchi S, Ku J, Hawtrey T, De Silvestro I, Malle E, Rees M, Thomas SR, Morris JC. Polyamine-Conjugated Nitroxides Are Efficacious Inhibitors of Oxidative Reactions Catalyzed by Endothelial-Localized Myeloperoxidase. Chem Res Toxicol 2021; 34:1681-1692. [PMID: 34085520 DOI: 10.1021/acs.chemrestox.1c00094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The heme enzyme myeloperoxidase (MPO) is a key mediator of endothelial dysfunction and a therapeutic target in cardiovascular disease. During inflammation, MPO released by circulating leukocytes is internalized by endothelial cells and transcytosed into the subendothelial extracellular matrix of diseased vessels. At this site, MPO mediates endothelial dysfunction by catalytically consuming nitric oxide (NO) and producing reactive oxidants, hypochlorous acid (HOCl) and the nitrogen dioxide radical (•NO2). Accordingly, there is interest in developing MPO inhibitors that effectively target endothelial-localized MPO. Here we studied a series of piperidine nitroxides conjugated to polyamine moieties as novel endothelial-targeted MPO inhibitors. Electron paramagnetic resonance analysis of cell lysates showed that polyamine conjugated nitroxides were efficiently internalized into endothelial cells in a heparan sulfate dependent manner. Nitroxides effectively inhibited the consumption of MPO's substrate hydrogen peroxide (H2O2) and formation of HOCl catalyzed by endothelial-localized MPO, with their efficacy dependent on both nitroxide and conjugated-polyamine structure. Nitroxides also differentially inhibited protein nitration catalyzed by both purified and endothelial-localized MPO, which was dependent on •NO2 scavenging rather than MPO inhibition. Finally, nitroxides uniformly inhibited the catalytic consumption of NO by MPO in human plasma. These studies show for the first time that nitroxides effectively inhibit local oxidative reactions catalyzed by endothelial-localized MPO. Novel polyamine-conjugated nitroxides, ethylenediamine-TEMPO and putrescine-TEMPO, emerged as efficacious nitroxides uniquely exhibiting high endothelial cell uptake and efficient inhibition of MPO-catalyzed HOCl production, protein nitration, and NO oxidation. Polyamine-conjugated nitroxides represent a versatile class of antioxidant drugs capable of targeting endothelial-localized MPO during vascular inflammation.
Collapse
Affiliation(s)
- Sophie Maiocchi
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia.,Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States.,School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jacqueline Ku
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Tom Hawtrey
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Irene De Silvestro
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Ernst Malle
- Gottfried Schatz Research Center, Molecular Biology & Biochemistry, Medical University of Graz, 8036 Graz, Austria
| | - Martin Rees
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Shane R Thomas
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jonathan C Morris
- School of Chemistry, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
14
|
Kargapolova Y, Geißen S, Zheng R, Baldus S, Winkels H, Adam M. The Enzymatic and Non-Enzymatic Function of Myeloperoxidase (MPO) in Inflammatory Communication. Antioxidants (Basel) 2021; 10:antiox10040562. [PMID: 33916434 PMCID: PMC8066882 DOI: 10.3390/antiox10040562] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Myeloperoxidase is a signature enzyme of polymorphonuclear neutrophils in mice and humans. Being a component of circulating white blood cells, myeloperoxidase plays multiple roles in various organs and tissues and facilitates their crosstalk. Here, we describe the current knowledge on the tissue- and lineage-specific expression of myeloperoxidase, its well-studied enzymatic activity and incoherently understood non-enzymatic role in various cell types and tissues. Further, we elaborate on Myeloperoxidase (MPO) in the complex context of cardiovascular disease, innate and autoimmune response, development and progression of cancer and neurodegenerative diseases.
Collapse
|
15
|
Thai T, Zhong F, Dang L, Chan E, Ku J, Malle E, Geczy CL, Keaney JF, Thomas SR. Endothelial-transcytosed myeloperoxidase activates endothelial nitric oxide synthase via a phospholipase C-dependent calcium signaling pathway. Free Radic Biol Med 2021; 166:255-264. [PMID: 33539947 PMCID: PMC10686581 DOI: 10.1016/j.freeradbiomed.2020.12.448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022]
Abstract
During vascular inflammation, the leukocyte-derived enzyme myeloperoxidase (MPO) is transcytosed across the endothelium and into the sub-endothelial extracellular matrix, where it promotes endothelial dysfunction by catalytically consuming nitric oxide (NO) produced by endothelial NO synthase (eNOS). In the presence of chloride ions and hydrogen peroxide (H2O2), MPO forms the oxidant hypochlorous acid (HOCl). Here we examined the short-term implications of HOCl produced by endothelial-transcytosed MPO for eNOS activity. Incubation of MPO with cultured aortic endothelial cells (ECs) resulted in its transport into the sub-endothelium. Exposure of MPO-containing ECs to low micromolar concentrations of H2O2 yielded enhanced rates of H2O2 consumption that correlated with HOCl formation and increased eNOS enzyme activity. The MPO-dependent activation of eNOS occurred despite reduced cellular uptake of the eNOS substrate l-arginine, which involved a decrease in the maximal activity (Vmax), but not substrate affinity (Km), of the major endothelial l-arginine transporter, cationic amino acid transporter-1. Activation of eNOS in MPO-containing ECs exposed to H2O2 involved a rapid elevation in cytosolic calcium and increased eNOS phosphorylation at Ser-1179 and de-phosphorylation at Thr-497. These signaling events were attenuated by intracellular calcium chelation, removal of extracellular calcium and inhibition of phospholipase C. This study shows that stimulation of endothelial-transcytosed MPO activates eNOS by promoting phospholipase C-dependent calcium signaling and altered eNOS phosphorylation at Ser-1179 and Thr-497. This may constitute a compensatory signaling response of ECs aimed at maintaining eNOS activity and NO production in the face of MPO-catalyzed oxidative stress.
Collapse
Affiliation(s)
- Thuan Thai
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; School of Education, University of Notre Dame Australia, Sydney, NSW, Australia
| | - Fei Zhong
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Lei Dang
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Enoch Chan
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Jacqueline Ku
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Ernst Malle
- Gottfried Schatz Research Center, Division of Molecular Biology & Biochemistry, Medical University of Graz, Graz, Austria
| | - Carolyn L Geczy
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - John F Keaney
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Shane R Thomas
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Yang Y, Liu L, Guo Z, Li L, Shao Y, Song M, Sun B. Investigation and assessment of neutrophil dysfunction early after severe burn injury. Burns 2021; 47:1851-1862. [PMID: 33820675 DOI: 10.1016/j.burns.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/09/2021] [Accepted: 02/11/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Extensive burn injury results in a complex immune response that is associated with mortality and prognosis. Studies on acquired immune and the development of sepsis in burn patients have been reported. However, one of the main cells in innate immune, neutrophil dysfunction in the burn shock stage has not been thoroughly characterized. METHODS Neutrophil chemotaxis, expression of neutrophil surface markers (P2X1 receptor, [P2RX1]), degranulation (myeloperoxidase [MPO], heparin-binding protein [HBP], matrix metalloproteinase-9 [MMP-9] and neutrophil elastase [NE]), oxidative burst capacity, neutrophil extracellular trap (NET) generation, phagocytosis and apoptosis were measured in 18 patients with major burns (≥30% total body surface area [TBSA]) within 48 h after burn injury. In addition, circulating neutrophils and vascular permeability in mice model with 30% TBSA third-degree burns were also observed and investigated. RESULTS Neutrophil functions were reduced considerably in burn shock stage, which was characterized by decreased chemotaxis, phagocytosis and abnormal bactericidal function. Increased release of heparin-binding protein (HBP) and the expression of P2RX1 on the neutrophil surface are related to fluid leakage and decreased chemotaxis during burn shock stage, respectively. The combination of HBP concentration in plasma and P2RX1 expression on neutrophils gives a better prediction of neutrophil dysfunction in burn-injured patients. CONCLUSION Neutrophil dysfunction plays a key role in the development of burn injury. Targeting the restoration of neutrophil function may be a feasible therapeutic intervention to help reduce fluid loss during shock and the severity of subsequent infection.
Collapse
Affiliation(s)
- Yunxi Yang
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Lu Liu
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Zaiwen Guo
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Linbin Li
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Mingming Song
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China.
| |
Collapse
|
17
|
Myeloperoxidase: Mechanisms, reactions and inhibition as a therapeutic strategy in inflammatory diseases. Pharmacol Ther 2021; 218:107685. [DOI: 10.1016/j.pharmthera.2020.107685] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022]
|
18
|
Kolářová H, Víteček J, Černá A, Černík M, Přibyl J, Skládal P, Potěšil D, Ihnatová I, Zdráhal Z, Hampl A, Klinke A, Kubala L. Myeloperoxidase mediated alteration of endothelial function is dependent on its cationic charge. Free Radic Biol Med 2021; 162:14-26. [PMID: 33271281 DOI: 10.1016/j.freeradbiomed.2020.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022]
Abstract
Endothelial cell (EC) glycocalyx (GLX) comprise a multicomponent layer of proteoglycans and glycoproteins. Alteration of its integrity contributes to chronic vascular inflammation and leads to the development of cardiovascular diseases. Myeloperoxidase (MPO), a highly abundant enzyme released by polymorphonuclear neutrophils, binds to the GLX and deleteriously affects vascular EC functions. The focus of this study was to elucidate the mechanisms of MPO-mediated alteration of GLX molecules, and to unravel subsequent changes in endothelial integrity and function. MPO binding to GLX of human ECs and subsequent internalization was mediated by cell surface heparan sulfate chains. Moreover, interaction of MPO, which is carrying a cationic charge, with anionic glycosaminoglycans (GAGs) resulted in reduction of their relative charge. By means of micro-viscometry and atomic force microscopy, we disclosed that MPO can crosslink GAG chains. MPO-dependent modulation of GLX structure was further supported by alteration of wheat germ agglutinin staining. Increased expression of ICAM-1 documented endothelial cell activation by both catalytically active and also inactive MPO. Furthermore, MPO increased vascular permeability connected with reorganization of intracellular junctions, however, this was dependent on MPO's catalytic activity. Novel proteins interacting with MPO during transcytosis were identified by proteomic analysis. Altogether, these findings provide evidence that MPO through interaction with GAGs modulates overall charge of the GLX, causing modification of its structure and thus affecting EC function. Importantly, our results also suggest a number of proteins interacting with MPO that possess a variety of cellular localizations and functions.
Collapse
Affiliation(s)
- Hana Kolářová
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Jan Víteček
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Anna Černá
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Marek Černík
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Jan Přibyl
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - Petr Skládal
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - David Potěšil
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Ivana Ihnatová
- Institute of Biostatistics and Analyses, Masaryk University, Kamenice 3, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Aleš Hampl
- Faculty of Medicine, Department of Histology and Embryology, Masaryk University, Kamenice 3, 625 00, Brno, Czech Republic
| | - Anna Klinke
- Clinic of General and Interventional Cardiology/Angiology, Agnes Wittenborg Institute of Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Lukáš Kubala
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic.
| |
Collapse
|
19
|
Bushueva OY. Genetic Variants rs1049255 CYBA and rs2333227 MPO are Associated with Susceptibility to Coronary Artery Disease in Russian Residents of Central Russia. KARDIOLOGIIA 2020; 60:1229. [PMID: 33228505 DOI: 10.18087/cardio.2020.10.n1229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Aim To study association of single-nucleotide polymorphisms rs1049255 CYBA and rs2333227 MPO with development of ischemic heart disease (IHD) in Russian residents of Central Russia.Material and methods The study material was DNA samples from 436 patients with IHD (265 men, 171 women; mean age, 61 years) and 370 sex- and age-matched arbitrarily healthy volunteers (209 men, 161 women; mean age, 60 years). Genotyping was performed by allelic discrimination with TaqMan probes.Results Comparative analysis of genotype frequency (log-additive regression model) showed that SNP rs1049255 CYBA (odds ratio, OR, 0.79 at 95 % confidence interval, CI, from 0.65 to 0.96; p=0.02) and rs2333227 MPO (OR 0.72 at 95 % CI from 0.55 to 0.95; p=0.02) were associated with a decreased risk of IHD adjusted for sex and age. Analysis of sex-specific effects showed that the protective effect of rs1049255 CYBA was evident only in men (OR 0.72 at 95 % CI from 0.55 to 0.94; p=0.16).Conclusion The study demonstrated a protective effect of rs1049255 CYBA and rs2333227 MPO with respect of IHD in Russians. The protective effect of rs1049255 CYBA was observed only in men.
Collapse
|
20
|
Myeloperoxidase: A versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther 2020; 221:107711. [PMID: 33137376 DOI: 10.1016/j.pharmthera.2020.107711] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Myeloperoxidase (MPO) is a prominent mammalian heme peroxidase and a fundamental component of the innate immune response against microbial pathogens. In recent times, MPO has received considerable attention as a key oxidative enzyme capable of impairing the bioactivity of nitric oxide (NO) and promoting endothelial dysfunction; a clinically relevant event that manifests throughout the development of inflammatory cardiovascular disease. Increasing evidence indicates that during cardiovascular disease, MPO is released intravascularly by activated leukocytes resulting in its transport and sequestration within the vascular endothelium. At this site, MPO catalyzes various oxidative reactions that are capable of promoting vascular inflammation and impairing NO bioactivity and endothelial function. In particular, MPO catalyzes the production of the potent oxidant hypochlorous acid (HOCl) and the catalytic consumption of NO via the enzyme's NO oxidase activity. An emerging paradigm is the ability of MPO to also influence endothelial function via non-catalytic, cytokine-like activities. In this review article we discuss the implications of our increasing knowledge of the versatility of MPO's actions as a mediator of cardiovascular disease and endothelial dysfunction for the development of new pharmacological agents capable of effectively combating MPO's pathogenic activities. More specifically, we will (i) discuss the various transport mechanisms by which MPO accumulates into the endothelium of inflamed or diseased arteries, (ii) detail the clinical and basic scientific evidence identifying MPO as a significant cause of endothelial dysfunction and cardiovascular disease, (iii) provide an up-to-date coverage on the different oxidative mechanisms by which MPO can impair endothelial function during cardiovascular disease including an evaluation of the contributions of MPO-catalyzed HOCl production and NO oxidation, and (iv) outline the novel non-enzymatic mechanisms of MPO and their potential contribution to endothelial dysfunction. Finally, we deliver a detailed appraisal of the different pharmacological strategies available for targeting the catalytic and non-catalytic modes-of-action of MPO in order to protect against endothelial dysfunction in cardiovascular disease.
Collapse
|
21
|
Alexandrovski M, Suciu S, Alexandrovski J. Joint Measurements of Leukocyte Elastase and Myeloperoxidase Promote Identification of the State of Neutrophils in Diabetic Patients. Biores Open Access 2020; 9:190-197. [PMID: 32908806 PMCID: PMC7473041 DOI: 10.1089/biores.2020.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 11/18/2022] Open
Abstract
The clinic of diabetes mellitus (DM) offers a number of hypotheses about the leading role of polymorphonuclear neutrophils (PMNs) in both oxidative stress and diabetic complications. However, the results of numerous studies are extremely controversial. Why is it so? We appreciated the clinical significance of simultaneous measurement data of several PMN parameters, which must complement each other. For this purpose, myeloperoxidase (MPO) and elastase (EL) were jointly analyzed in the blood plasma from 160 type 2 diabetes mellitus patients with high levels of HbA1c. A weakly positive correlation (r ∼ 0.56) was observed between MPO and EL analytical data, and any correlation between the concentrations of MPO/EL and HbA1c was absent. Medians of 160 measurements of MPO/EL concentrations were ∼103/190 ng/mL, and 95% of all results were in the range below 320/1016 ng/mL, respectively. The share of DM patients whose concentrations of MPO, EL, or either of two parameters exceeded the corresponding reference values was 65%, 80%, and 82.5%, respectively. These findings—a high intensity of neutrophil degranulation process—indicated that some diabetic conditions promote the transfer of PMNs to an “arousal” or “subactivation” state, which is identical or similar to their activation, providing in vivo an almost inexhaustible source of extremely “aggressive” MPO and EL. Thus, the conjoint MPO/EL measurements confirm the leading role of PMNs in the development of various complications of diabetes. The paradox is that the diagnostic significance of MPO/EL as independent parameters in diabetic patients is unambiguous for a number of reasons.
Collapse
Affiliation(s)
| | - Soimita Suciu
- Department of Physiology, Cluj-Napoca University of Medicine and Pharmacy, Cluj, Romania
| | | |
Collapse
|
22
|
Abstract
Normal pregnancy is a complex and dynamic process that requires significant adaptation from the maternal system. Failure of this adaptive process in pregnancy contributes to many pregnancy related disorders, including the hypertensive disorders of pregnancy. This article discusses placental development and how abnormalities in the process of vascular remodeling contribute to the multisystem maternal and fetal disease that is preeclampsia and fetal growth restriction. We review some of the consequences of this condition on the mother and fetus, aspects of the clinical management of preeclampsia and how it can influence both mother and infant in the postnatal period and beyond.
Collapse
|
23
|
Blood Levels of Free-Circulating Mitochondrial DNA in Septic Shock and Postsurgical Systemic Inflammation and Its Influence on Coagulation: A Secondary Analysis of a Prospective Observational Study. J Clin Med 2020; 9:jcm9072056. [PMID: 32629885 PMCID: PMC7408641 DOI: 10.3390/jcm9072056] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/29/2022] Open
Abstract
Major surgery is regularly associated with clinical signs of systemic inflammation, which potentially affects the rapid identification of sepsis. Therefore, this secondary analysis of an observational study aims to determine whether NADH dehydrogenase 1 (ND1) mitochondrial DNA (mtDNA) could be used as a potential biomarker for the discrimination between septic shock and postsurgical systemic inflammation. Overall, 80 patients were included (septic shock (n = 20), cardiac artery bypass grafting (CABG, n = 20), major abdominal surgery (MAS, n = 20), and matched controls (CTRL, n = 20)). Quantitative PCR was performed to measure ND1 mtDNA. Thromboelastography was used to analyze the coagulatory system. Free-circulating ND1 mtDNA levels were significantly higher in septic shock patients compared to patients suffering from post-surgical inflammation ({copies/µL}: CTRL: 1208 (668-2685); septic shock: 3823 (2170-7318); CABG: 1272 (417-2720); and MAS: 1356 (694-2845); CTRL vs. septic shock: p < 0.001; septic shock vs. CABG: p < 0.001; septic shock vs. MAS: p = 0.006; CABG vs. MAS: p = 0.01). ND1 mtDNA levels in CABG patients showed a strong positive correlation with fibrinogen (correlation coefficient [r]= 0.57, p < 0.001) and fibrinogen-dependent thromboelastographic assays (maximum clot firmness, EXTEM: r = 0.35, p = 0.01; INTEM: r = 0.31, p = 0.02; FIBTEM: r = 0.46, p < 0.001). In conclusion, plasma levels of free-circulating ND1 mtDNA were increased in septic shock patients and were discriminative between sepsis and surgery-induced inflammation. Furthermore, this study showed an association between ND1 mtDNA and a fibrinogen-dependent pro-coagulatory shift in cardiac surgical patients.
Collapse
|
24
|
Davies MJ, Hawkins CL. The Role of Myeloperoxidase in Biomolecule Modification, Chronic Inflammation, and Disease. Antioxid Redox Signal 2020; 32:957-981. [PMID: 31989833 DOI: 10.1089/ars.2020.8030] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Significance: The release of myeloperoxidase (MPO) by activated leukocytes is critical in innate immune responses. MPO produces hypochlorous acid (HOCl) and other strong oxidants, which kill bacteria and other invading pathogens. However, MPO also drives the development of numerous chronic inflammatory pathologies, including atherosclerosis, neurodegenerative disease, lung disease, arthritis, cancer, and kidney disease, which are globally responsible for significant patient mortality and morbidity. Recent Advances: The development of imaging approaches to precisely identify the localization of MPO and the molecular targets of HOCl in vivo is an important advance, as typically the involvement of MPO in inflammatory disease has been inferred by its presence, together with the detection of biomarkers of HOCl, in biological fluids or diseased tissues. This will provide valuable information in regard to the cell types responsible for releasing MPO in vivo, together with new insight into potential therapeutic opportunities. Critical Issues: Although there is little doubt as to the value of MPO inhibition as a protective strategy to mitigate tissue damage during chronic inflammation in experimental models, the impact of long-term inhibition of MPO as a therapeutic strategy for human disease remains uncertain, in light of the potential effects on innate immunity. Future Directions: The development of more targeted MPO inhibitors or a treatment regimen designed to reduce MPO-associated host tissue damage without compromising pathogen killing by the innate immune system is therefore an important future direction. Similarly, a partial MPO inhibition strategy may be sufficient to maintain adequate bacterial activity while decreasing the propagation of inflammatory pathologies.
Collapse
Affiliation(s)
- Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
25
|
McLaughlin K, Nadeem L, Wat J, Baczyk D, Lye SJ, Kingdom JC. Low molecular weight heparin promotes transcription and release of placental growth factor from endothelial cells. Am J Physiol Heart Circ Physiol 2020; 318:H1008-H1017. [PMID: 32196359 DOI: 10.1152/ajpheart.00109.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Circulating levels of placental growth factor (PlGF) are significantly reduced in women who develop preeclampsia. Low molecular weight heparin (LMWH) has been shown to acutely elevate circulating PlGF levels in pregnant women at increased risk of preeclampsia. The objective of the current investigation was to determine the mechanisms by which LMWH mediates the extracellular release of PlGF from endothelial cells. Cultured human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs) were exposed to LMWH; PlGF transcription, translation, mobilization, and secretion were then assessed. LMWH significantly increased the release of PlGF from both HAECs and HUVECs. LMWH treatment promoted a significant increase of PlGF-1 mRNA expression in HAECs, accompanied by the intracellular transport and release of PlGF into the conditioned media. LMWH-mediated release of PlGF from HAECs was not directly mediated by extracellular mobilization, synthesis, or stability of PlGF mRNA/protein. LMWH exposure promotes the release of PlGF from endothelial cells through the upregulation of PlGF-1 mRNA expression. Stimulation of circulating PlGF levels by LMWH may be an important mechanism by which LMWH could reduce the risk of preeclampsia or minimize disease severity.NEW & NOTEWORTHY There are few therapeutic options available for the prevention of preeclampsia, a serious hypertensive disorder of pregnancy. Women who subsequently develop preeclampsia exhibit significantly reduced circulating levels of the proangiogenic placental growth factor protein. Low molecular weight heparin (LMWH) has previously been investigated as a preventative therapy against the development of preeclampsia; however, its mechanism of action is not known. The current study determined that LMWH promotes the transcription and release of placental growth factor protein from endothelial cells, providing a mechanistic basis by which LMWH could reduce the risk of preeclampsia or minimize disease severity.
Collapse
Affiliation(s)
- Kelsey McLaughlin
- Division of Cardiology, Department of Medicine, Sinai Health System, University of Toronto, Toronto, Canada.,The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Lubna Nadeem
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Jovian Wat
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Dora Baczyk
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Stephen J Lye
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Sinai Health System, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - John C Kingdom
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada.,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Sinai Health System, University of Toronto, Toronto, Canada
| |
Collapse
|
26
|
Binding of myeloperoxidase to the extracellular matrix of smooth muscle cells and subsequent matrix modification. Sci Rep 2020; 10:666. [PMID: 31959784 PMCID: PMC6971288 DOI: 10.1038/s41598-019-57299-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/18/2019] [Indexed: 11/08/2022] Open
Abstract
The extracellular matrix (ECM) of tissues is susceptible to modification by inflammation-associated oxidants. Considerable data support a role for hypochlorous acid (HOCl), generated by the leukocyte-derived heme-protein myeloperoxidase (MPO) in these changes. HOCl can modify isolated ECM proteins and cell-derived matrix, with this resulting in decreased cell adhesion, modulated proliferation and gene expression, and phenotypic changes. Whether this arises from free HOCl, or via site-specific reactions is unresolved. Here we examine the mechanisms of MPO-mediated changes to human coronary smooth muscle cell ECM. MPO is shown to co-localize with matrix fibronectin as detected by confocal microscopy, and bound active MPO can initiate ECM modification, as detected by decreased antibody recognition of fibronectin, versican and type IV collagen, and formation of protein carbonyls and HOCl-mediated damage. These changes are recapitulated by a glucose/glucose oxidase/MPO system where low continuous fluxes of H2O2 are generated. HOCl-induced modifications enhance MPO binding to ECM proteins as detected by ELISA and MPO activity measurements. These data demonstrate that MPO-generated HOCl induces ECM modification by interacting with ECM proteins in a site-specific manner, and generates alterations that increase MPO adhesion. This is proposed to give rise to an increasing cycle of alterations that contribute to tissue damage.
Collapse
|
27
|
Flow Cytometry-Based Quantification of Neutrophil Extracellular Traps Shows an Association with Hypercoagulation in Septic Shock and Hypocoagulation in Postsurgical Systemic Inflammation-A Proof-of-Concept Study. J Clin Med 2020; 9:jcm9010174. [PMID: 31936385 PMCID: PMC7019434 DOI: 10.3390/jcm9010174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 12/23/2022] Open
Abstract
This proof-of-concept study aimed to evaluate a novel method of flow cytometry-based quantification of neutrophil extracellular traps (NETs) in septic shock patients and to identify possible interactions between the number of free-circulating NETs and alterations of the coagulatory system. Patients suffering from septic shock, a matched control group (CTRL), and patients suffering from systemic inflammation after cardiac (CABG) or major abdominal surgery (MAS) were enrolled in this prospective proof-of-concept study. Compared to the matched controls, free-circulating NETs were significantly elevated in septic shock and postsurgical patients (data are presented in median (IQR)); septic shock: (2.7 (1.9-3.9); CABG: 2.7 (2.1-3.7); MAS: 2.7 (2.1-3.9); CTRL: 1.6 (1-2); CTRL vs. septic shock: p = 0.001; CTRL vs. CABG: p < 0.001; CTRL vs. MAS: p < 0.001). NETs correlated positively with FIBTEM mean clot firmness (MCF) in septic shock patients (r = 0.37, p < 0.01) while they correlated negatively in surgical patients (CABG: r = -0.28, p < 0.01; MAS: r = -0.25, p = 0.03). Flow-cytometric quantification of NETs showed a significant increase in free-circulating NETs under inflammatory conditions. Furthermore, this study hints to an association of the number of NETs with hypercoagulation in septic shock patients and hypocoagulation in surgery-induced inflammation.
Collapse
|
28
|
The Peroxisome Proliferator-Activated Receptor γ Agonist Pioglitazone Protects Vascular Endothelial Function in Hypercholesterolemic Rats by Inhibiting Myeloperoxidase. Cardiol Res Pract 2020; 2020:1845969. [PMID: 32190383 PMCID: PMC7063881 DOI: 10.1155/2020/1845969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Hypercholesterolemia- (HC-) induced endothelial dysfunction is the first step of atherogenesis, and the peroxisome proliferator-activated receptor γ (PPARγ (PPARγ (PPARγ) has been reported to attenuate atherosclerosis formation; however, the underlying mechanisms are not fully understood. The present study was designed to determine whether myeloperoxidase (MPO) mediates HC-induced endothelial dysfunction and the role of the PPARγ agonist pioglitazone (PIO) in attenuating endothelial dysfunction. Methods Male Wistar rats were fed with normal or high cholesterol diets for 8 weeks. HC rats were randomized to receive dapsone (DDS, the MPO inhibitor) during the last 6 days or PIO for the remaining 4 weeks. Vascular endothelial function was determined by comparing vasorelaxation to ACh, an endothelium-dependent vasodilator, and SNP, an endothelium-independent vasodilator in vascular rings in vitro. The vascular MPO activity, NOx content, and cGMP level were measured by the MPO activity assay kit, NO assay kit, and cGMP RIA kit. Results Compared with rats fed with normal diet, endothelium-dependent vasodilation, NOx content, and cGMP level were decreased, and MPO activity was increased in thoracic aortas of rats fed with HC diet. There was a negative correlation between vascular endothelial function, NOx content or cGMP level, and MPO activity. PIO obviously reduced the MPO activity, increased NOx content and cGMP level, and improved endothelium-dependent vasodilation function in HC rats, which was essentially the same as that seen with DDS. And, there was a negative correlation between vascular endothelial function, NOx content or cGMP level, and MPO activity in the HC group and the PIO intervention group. Conclusion MPO might provoke vascular endothelial dysfunction in hypercholesterolemic rats by reducing the NO biological activity and impairing the NO/cGMP/cGK signaling pathway. PIO might inhibit vascular MPO activity and increase NO bioavailability with the net result of reversing endothelial dysfunction.
Collapse
|
29
|
Correa S, Pena-Esparragoza JK, Scovner KM, Waikar SS, Mc Causland FR. Myeloperoxidase and the Risk of CKD Progression, Cardiovascular Disease, and Death in the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis 2019; 76:32-41. [PMID: 31864821 DOI: 10.1053/j.ajkd.2019.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/07/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Myeloperoxidase (MPO) catalyzes the formation of reactive nitrogen species and levels are elevated in patients with chronic kidney disease (CKD). Although increased oxidative stress and inflammation are associated with progression of CKD and cardiovascular disease (CVD), relationships between MPO concentration, CKD progression, CVD, and death remain unclear. STUDY DESIGN Prospective cohort. SETTING & PARTICIPANTS 3,872 participants from the Chronic Renal Insufficiency Cohort (CRIC) who had MPO measured at baseline. EXPOSURE Baseline MPO concentration. OUTCOMES CKD progression (kidney transplantation, dialysis initiation, or 50% decline in baseline estimated glomerular filtration rate [eGFR] and eGFR≤15mL/min/1.73m2), CVD (heart failure, myocardial infarction, or stroke), and death. ANALYTICAL APPROACH Cox proportional hazards models. RESULTS In adjusted analyses, higher MPO level (per 1-SD increase in log-transformed MPO) was associated with 10% higher risk for CKD progression (adjusted HR, 1.10; 95% CI, 1.01-1.19; P=0.03), 12% higher risk for CVD (adjusted HR, 1.12; 95% CI, 1.03-1.22; P<0.01), and 13% increased risk for death (adjusted HR, 1.13; 95% CI, 1.04-1.22; P<0.01). There was evidence for effect modification of the association of MPO level with CKD progression by baseline eGFR (P interaction=0.02), but not for CVD (P interaction=0.2) or death (P interaction=0.1). In stratified analyses, MPO level (per 1-SD increase in log-transformed MPO) was associated with greater risk for CKD progression among participants with eGFR>45mL/min/1.73m2 (adjusted HR, 1.23; 95% CI, 1.03-1.46; P=0.02) compared with those with eGFR≤45mL/min/1.73m2 (adjusted HR, 1.10; 95% CI, 1.02-1.20; P=0.02). The association of MPO level with CVD and death was no longer significant after adjustment for cardiac biomarkers. LIMITATIONS Potential residual confounding, lack of repeated measurements of MPO. CONCLUSIONS Higher MPO level was associated with increased risk for CKD progression, but not with CVD and death in patients with CKD from CRIC. Whether therapies aimed at reducing MPO activity can result in improved clinical outcomes is yet to be determined.
Collapse
Affiliation(s)
- Simon Correa
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| | | | - Katherine M Scovner
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Sushrut S Waikar
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA
| | - Finnian R Mc Causland
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Iodide modulates protein damage induced by the inflammation-associated heme enzyme myeloperoxidase. Redox Biol 2019; 28:101331. [PMID: 31568923 PMCID: PMC6812061 DOI: 10.1016/j.redox.2019.101331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 01/15/2023] Open
Abstract
Iodide ions (I-) are an essential dietary mineral, and crucial for mental and physical development, fertility and thyroid function. I- is also a high affinity substrate for the heme enzyme myeloperoxidase (MPO), which is involved in bacterial cell killing during the immune response, and also host tissue damage during inflammation. In the presence of H2O2 and Cl-, MPO generates the powerful oxidant hypochlorous acid (HOCl), with excessive formation of this species linked to multiple inflammatory diseases. In this study, we have examined the hypothesis that elevated levels of I- would decrease HOCl formation and thereby protein damage induced by a MPO/Cl-/H2O2 system, by acting as a competitive substrate. The presence of increasing I- concentrations (0.1-10 μM; i.e. within the range readily achievable by oral supplementation in humans), decreased damage to both model proteins and extracellular matrix components as assessed by gross structural changes (SDS-PAGE), antibody recognition of parent and modified protein epitopes (ELISA), and quantification of both parent amino acid loss (UPLC) and formation of the HOCl-biomarker 3-chlorotyrosine (LC-MS) (reduced by ca. 50% at 10 μM I-). Elevated levels of I- ( > 1 μM) also protected against functional changes as assessed by a decreased loss of adhesion (eg. 40% vs. < 22% with >1 μM I-) of primary human coronary artery endothelial cells (HCAECs), to MPO-modified human plasma fibronectin. These data indicate that low micromolar concentrations of I-, which can be readily achieved in humans and are readily tolerated, may afford protection against cell and tissue damage induced by MPO.
Collapse
|
31
|
Manchanda K, Kolarova H, Kerkenpaß C, Mollenhauer M, Vitecek J, Rudolph V, Kubala L, Baldus S, Adam M, Klinke A. MPO (Myeloperoxidase) Reduces Endothelial Glycocalyx Thickness Dependent on Its Cationic Charge. Arterioscler Thromb Vasc Biol 2019; 38:1859-1867. [PMID: 29903730 DOI: 10.1161/atvbaha.118.311143] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective- The leukocyte heme-enzyme MPO (myeloperoxidase) exerts proinflammatory effects on the vascular system primarily linked to its catalytic properties. Recent studies have shown that MPO, depending on its cationic charge, mediates neutrophil recruitment and activation. Here, we further investigated MPO's extracatalytic properties and its effect on endothelial glycocalyx (EG) integrity. Approach and Results- In vivo staining of murine cremaster muscle vessels with Alcian Blue 8GX provided evidence of an MPO-dependent decrease in anionic charge of the EG. MPO binding to the glycocalyx was further characterized using Chinese hamster ovary cells and its glycosaminoglycan mutants-pgsA-745 (mutant Chinese hamster ovary cells lacking heparan sulfate and chondroitin sulfate glycosaminoglycan) and pgsD-677 (mutant Chinese hamster ovary cells lacking heparan sulfate glycosaminoglycan), which revealed heparan sulfate as the main mediator of MPO binding. Further, EG integrity was assessed in terms of thickness using intravital microscopy of murine cremaster muscle. A significant reduction in EG thickness was observed on infusion of catalytically active MPO, as well as mutant inactive MPO and cationic polymer polylysine. Similar effects were also observed in wild-type mice after a local inflammatory stimulus but not in MPO-knockout mice. The reduction in EG thickness was reversed after removal of vessel-bound MPO, suggesting a possible physical collapse of the EG. Last, experiments with in vivo neutrophil depletion revealed that MPO also induced neutrophil-mediated shedding of the EG core protein, Sdc1 (syndecan-1). Conclusions- These findings provide evidence that MPO, via ionic interaction with heparan sulfate side chains, can cause neutrophil-dependent Sdc1 shedding and collapse of the EG structure.
Collapse
Affiliation(s)
- Kashish Manchanda
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Cologne Cardiovascular Research Center, University of Cologne, Germany (K.M., S.B., A.K.)
| | - Hana Kolarova
- Institute of Biophysics AS CR, Brno, Czech Republic (H.K., J.V., L.K.)
- International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic (H.K., J.V., L.K., A.K.)
| | - Christina Kerkenpaß
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
| | - Martin Mollenhauer
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
| | - Jan Vitecek
- Institute of Biophysics AS CR, Brno, Czech Republic (H.K., J.V., L.K.)
- International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic (H.K., J.V., L.K., A.K.)
| | - Volker Rudolph
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
| | - Lukas Kubala
- Institute of Biophysics AS CR, Brno, Czech Republic (H.K., J.V., L.K.)
- International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic (H.K., J.V., L.K., A.K.)
| | - Stephan Baldus
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Cologne Cardiovascular Research Center, University of Cologne, Germany (K.M., S.B., A.K.)
| | - Matti Adam
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
| | - Anna Klinke
- From the Department of Cardiology, Heart Center, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- Center for Molecular Medicine Cologne, University of Cologne, Germany (K.M., C.K., M.M., V.R., S.B., M.A., A.K.)
- International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic (H.K., J.V., L.K., A.K.)
- Cologne Cardiovascular Research Center, University of Cologne, Germany (K.M., S.B., A.K.)
| |
Collapse
|
32
|
Ndrepepa G. Myeloperoxidase - A bridge linking inflammation and oxidative stress with cardiovascular disease. Clin Chim Acta 2019; 493:36-51. [PMID: 30797769 DOI: 10.1016/j.cca.2019.02.022] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/17/2022]
Abstract
Myeloperoxidase (MPO) is a member of the superfamily of heme peroxidases that is mainly expressed in neutrophils and monocytes. MPO-derived reactive species play a key role in neutrophil antimicrobial activity and human defense against various pathogens primarily by participating in phagocytosis. Elevated MPO levels in circulation are associated with inflammation and increased oxidative stress. Multiple lines of evidence suggest an association between MPO and cardiovascular disease (CVD) including coronary artery disease, congestive heart failure, arterial hypertension, pulmonary arterial hypertension, peripheral arterial disease, myocardial ischemia/reperfusion-related injury, stroke, cardiac arrhythmia and venous thrombosis. Elevated MPO levels are associated with a poor prognosis including increased risk for overall and CVD-related mortality. Elevated MPO may signify an increased risk for CVD for at least 2 reasons. First, low-grade inflammation and increased oxidative stress coexist with many metabolic abnormalities and comorbidities and consequently an elevated MPO level may represent an increased cardiometabolic risk in general. Second, MPO produces a large number of highly reactive species which can attack, destroy or modify the function of every known cellular component. The most common MPO actions relevant to CVD are generation of dysfunctional lipoproteins with an increased atherogenicity potential, reduced NO availability, endothelial dysfunction, impaired vasoreactivity and atherosclerotic plaque instability. These actions strongly suggest that MPO is directly involved in the pathophysiology of CVD. In this regard MPO may be seen as a mediator or an instrument through which inflammation promotes CVD at molecular and cellular level. Clinical value of MPO therapeutic inhibition remains to be tested.
Collapse
Affiliation(s)
- Gjin Ndrepepa
- Department of Adult Cardiology, Deutsches Herzzentrum München, Technische Universität, Lazarettstrasse 36, 80636 Munich, Germany.
| |
Collapse
|
33
|
Gan LM, Lagerström-Fermér M, Ericsson H, Nelander K, Lindstedt EL, Michaëlsson E, Kjaer M, Heijer M, Whatling C, Fuhr R. Safety, tolerability, pharmacokinetics and effect on serum uric acid of the myeloperoxidase inhibitor AZD4831 in a randomized, placebo-controlled, phase I study in healthy volunteers. Br J Clin Pharmacol 2019; 85:762-770. [PMID: 30618054 PMCID: PMC6422671 DOI: 10.1111/bcp.13855] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 12/11/2018] [Accepted: 12/24/2018] [Indexed: 11/28/2022] Open
Abstract
Aims Myeloperoxidase activity can contribute to impaired vascular endothelial function and fibrosis in chronic inflammation‐related cardiovascular disease. Here, we investigated the safety, tolerability and pharmacokinetics of the myeloperoxidase inhibitor, AZD4831. Methods In this randomized, single‐blind, placebo‐controlled, phase I, first‐in‐human study, healthy men in five sequential cohorts were randomized 3:1 to receive a single oral dose of AZD4831 (5, 15, 45, 135 or 405 mg) or placebo, after overnight fasting. After at least 7 days' washout, one cohort additionally received AZD4831 45 mg after a high‐calorie meal. Results Forty men participated in the study (eight per cohort: AZD4831, n = 6; placebo, n = 2). AZD4831 distributed rapidly into plasma, with a half‐life of 38.2–50.0 hours. The area under the plasma concentration–time curve (AUC) increased proportionally with dose (AUC0–∝ slope estimate 1.060; 95% confidence interval [CI] 0.9943, 1.127). Increases in maximum plasma concentration were slightly more than dose proportional (slope estimate 1.201; 95% CI 1.071, 1.332). Food intake reduced AZD4831 absorption rate but did not substantially affect overall exposure or plasma half‐life (n = 4). Serum uric acid concentrations decreased by 71.77 (95% CI 29.15, 114.39) and 84.42 (58.90, 109.94) μmol L−1 with AZD4831 135 mg and 405 mg, respectively. Maculopapular rash (moderate intensity) occurred in 4/30 participants receiving AZD4831 (13.3%). No other safety concerns were identified. Conclusions AZD4831 was generally well tolerated, rapidly absorbed, had a long plasma half‐life and lowered uric acid concentrations after single oral doses in healthy men. These findings support the further clinical development of AZD4831.
Collapse
Affiliation(s)
- Li-Ming Gan
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Hans Ericsson
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Karin Nelander
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Erik Michaëlsson
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Maria Heijer
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Carl Whatling
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rainard Fuhr
- PAREXEL Early Phase Clinical Unit, Berlin, Germany
| |
Collapse
|
34
|
Beck HC, Jensen LO, Gils C, Ilondo AMM, Frydland M, Hassager C, Møller-Helgestad OK, Møller JE, Rasmussen LM. Proteomic Discovery and Validation of the Confounding Effect of Heparin Administration on the Analysis of Candidate Cardiovascular Biomarkers. Clin Chem 2018; 64:1474-1484. [DOI: 10.1373/clinchem.2017.282665] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 07/05/2018] [Indexed: 12/13/2022]
Abstract
Abstract
BACKGROUND
Several plasma proteins have been suggested as markers for a variety of cardiovascular conditions but fail to qualify in independent patient cohorts. This may relate to interference of medication on plasma protein concentrations. We used proteomics to identify plasma proteins that changed in concentration with heparin administration and therefore potentially may confound their evaluation as biomarkers in situations in which heparin is used.
METHODS
We used a proteomic approach based on isobaric tagging and nano-LC-MS/MS analysis to quantify several hundred proteins in a discovery study in which individual plasma samples from 9 patients at intravascular ultrasound follow-up 12 months after an acute myocardial infarction before heparin administration and 2, 15, and 60 min after heparin administration; we validated our findings in 500 individual plasma samples obtained at admission from patients with suspected ST segment elevation myocardial infarction (STEMI), of whom 363 were treated with heparin before admission.
RESULTS
In the discovery study, 25 of 653 identified plasma proteins displayed a changed concentration after heparin administration (Bonferroni-corrected P value at P < 7.66 × 10−5). Fourteen of the proteins changed significantly among heparin-treated patients in the validation study (nominal significance level of P < 6.92 × 10−5). Among heparin-affected proteins in both the discovery study and the validation study were midkine, spondin 1, secreted frizzled-like protein 1, lipoprotein lipase, and follistatin, all previously associated with STEMI.
CONCLUSIONS
Medications such as heparin administration given before blood sampling may confound biomarker discovery and should be carefully considered in such studies.
Collapse
Affiliation(s)
- Hans C Beck
- Department of Clinical Biochemistry and Pharmacology
- Centre for Clinical Proteomics
| | - Lisette O Jensen
- Department of Cardiology
- Center for Individualized Medicine in Arterial Diseases, Odense University Hospital, University of Southern Denmark, Denmark
| | | | | | - Martin Frydland
- Heart Center, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | - Ole K Møller-Helgestad
- Department of Cardiology
- Center for Individualized Medicine in Arterial Diseases, Odense University Hospital, University of Southern Denmark, Denmark
| | - Jacob E Møller
- Department of Cardiology
- Center for Individualized Medicine in Arterial Diseases, Odense University Hospital, University of Southern Denmark, Denmark
| | - Lars M Rasmussen
- Department of Clinical Biochemistry and Pharmacology
- Centre for Clinical Proteomics
- Center for Individualized Medicine in Arterial Diseases, Odense University Hospital, University of Southern Denmark, Denmark
| |
Collapse
|
35
|
Wat JM, Audette MC, Kingdom JC. Molecular actions of heparin and their implications in preventing pre-eclampsia. J Thromb Haemost 2018; 16:S1538-7836(22)02212-7. [PMID: 29877031 DOI: 10.1111/jth.14191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Indexed: 12/17/2022]
Abstract
Pre-eclampsia, a hypertensive disorder of pregnancy, continues to be a significant cause of global maternal morbidity. Low-dose aspirin remains the only standard-of-care prophylactic therapy for preventing pre-eclampsia, but is limited in efficacy. Heparin and its derivatives may further enhance the efficacy of aspirin therapy to prevent pre-eclampsia, but the mechanisms mediating this augmentative effect are not known. Although heparin is an anticoagulant agent, it also possesses many anticoagulant-independent properties that may be relevant in the prevention of pre-eclampsia, including effects on placental, vascular and inflammatory function. This review summarizes the non-anticoagulant properties of heparin, and extrapolates how these actions may influence the trajectory of pre-eclampsia pathogenesis as a means of pathway-specific therapy.
Collapse
Affiliation(s)
- J M Wat
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - M C Audette
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - J C Kingdom
- Research Centre for Women's and Infant's Health, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Grigorieva DV, Gorudko IV, Kostevich VA, Vasilyev VB, Cherenkevich SN, Panasenko OM, Sokolov AV. [Exocytosis of myeloperoxidase from activated neutrophils in the presence of heparin]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:16-22. [PMID: 29460830 DOI: 10.18097/pbmc20186401016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Exocytosis of myeloperoxidase (MPO) from activated neutrophils in the presence of the anionic polysaccharide heparin was studied. It was determined that the optimal concentration of heparin (0.1 u/ml), at which there is no additional activation of cells (absence of amplification of exocytosis of lysozyme contained in specific and azurophilic granules). It was found that after preincubation of cells with heparin (0.1 u/ml) the exocytosis of MPO from neutrophils activated by various stimulants (fMLP, PMA, plant lectins CABA and PHA-L) increased compared to that under the action of activators alone. In addition, it was shown that heparin in the range of concentrations 0.1-50 u/ml did not affect on the peroxidase activity of the MPO isolated from leukocytes. Thus, the use of heparin at a concentration of 0.1 u/ml avoids the artifact caused by the "loss" of MPO in a result of its binding to neutrophils, and increases the accuracy of the method of registration the degranulation of azurophilic granules of neutrophils based on determination of the concentration or peroxidase activity of MPO in cell supernatants.
Collapse
Affiliation(s)
| | | | - V A Kostevich
- Institute for Experimental Medicine, Saint-Petersburg, Russia; Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
| | - V B Vasilyev
- Institute for Experimental Medicine, Saint-Petersburg, Russia; Saint Petersburg University, Saint-Petersburg, Russia
| | | | - O M Panasenko
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia
| | - A V Sokolov
- Institute for Experimental Medicine, Saint-Petersburg, Russia; Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia; Saint Petersburg University, Saint-Petersburg, Russia; Centre of Preclinical Translational Research, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| |
Collapse
|
37
|
McLaughlin K, Scholten RR, Parker JD, Ferrazzi E, Kingdom JCP. Low molecular weight heparin for the prevention of severe preeclampsia: where next? Br J Clin Pharmacol 2018; 84:673-678. [PMID: 29226532 DOI: 10.1111/bcp.13483] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/29/2017] [Indexed: 12/14/2022] Open
Abstract
Low molecular weight heparin has been extensively evaluated for the prevention of preeclampsia in high-risk pregnant women; however, the results from these trials have been conflicting. This review discusses the potential mechanisms of action of low molecular weight heparin for the prevention of severe preeclampsia, how to optimize the selection of high-risk women for participation in future trials, and the importance of trial standardization.
Collapse
Affiliation(s)
- Kelsey McLaughlin
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Canada.,Department of Medicine, Division of Cardiology, Sinai Health System, University of Toronto, Canada
| | - Ralph R Scholten
- Department of Obstetrics and Gynaecology, Division of Maternal-Fetal Medicine, Sinai Health System, University of Toronto, Canada
| | - John D Parker
- Department of Medicine, Division of Cardiology, Sinai Health System, University of Toronto, Canada
| | - Enrico Ferrazzi
- Department of Obstetrics and Gynaecology, Buzzi Children's Hospital, University of Milan, Italy
| | - John C P Kingdom
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Canada.,Department of Obstetrics and Gynaecology, Division of Maternal-Fetal Medicine, Sinai Health System, University of Toronto, Canada
| |
Collapse
|
38
|
McLaughlin K, Audette MC, Parker JD, Kingdom JC. Mechanisms and Clinical Significance of Endothelial Dysfunction in High-Risk Pregnancies. Can J Cardiol 2018; 34:371-380. [PMID: 29571421 DOI: 10.1016/j.cjca.2018.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 10/18/2022] Open
Abstract
The maternal cardiovascular system undergoes critical anatomic and functional adaptations to achieve a successful pregnancy outcome which, if disrupted, can result in complications that significantly affect maternal and fetal health. Complications that involve the maternal cardiovascular system are among the most common disorders of pregnancy, including gestational hypertension, preeclampsia, gestational diabetes, and impaired fetal growth. As a central feature, maternal endothelial dysfunction is hypothesized to play a predominant role in mediating the pathogenesis of these high-risk pregnancies, and as such, might proceed and precipitate the clinical presentation of these pregnancy disorders. Improving or normalizing maternal endothelial function in high-risk pregnancies might be an effective therapeutic strategy to ameliorate maternal and fetal clinical outcomes.
Collapse
Affiliation(s)
- Kelsey McLaughlin
- Department of Medicine, Division of Cardiology, Sinai Health System, University of Toronto, Toronto, Ontario, Canada; The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Melanie C Audette
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - John D Parker
- Department of Medicine, Division of Cardiology, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - John C Kingdom
- The Centre for Women's and Infant's Health at the Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, Division of Maternal-Fetal Medicine, Sinai Health System, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
39
|
Angiogenic Factor Profiles in Pregnant Women With a History of Early-Onset Severe Preeclampsia Receiving Low-Molecular-Weight Heparin Prophylaxis. Obstet Gynecol 2018; 131:63-69. [DOI: 10.1097/aog.0000000000002380] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
40
|
Cilostazol attenuates intimal hyperplasia in a mouse model of chronic kidney disease. PLoS One 2017; 12:e0187872. [PMID: 29206849 PMCID: PMC5716535 DOI: 10.1371/journal.pone.0187872] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 10/29/2017] [Indexed: 11/21/2022] Open
Abstract
Intimal hyperplasia (IH) is a common cause of vasculopathy due to direct endothelial damage (such as post-coronary revascularization) or indirect injury (such as chronic kidney disease, or CKD). Although the attenuation of coronary revascularization-induced IH (direct-vascular-injury-induced IH) by cilostazol, a phosphodiesterase III inhibitor, has been demonstrated, our understanding of the effect on CKD-induced IH (indirect-vascular-injury-induced IH) is limited. Herein, we tested if cilostazol attenuated CKD-induced IH in a mouse model of ischemic-reperfusion injury with unilateral nephrectomy (Chr I/R), a normotensive non-proteinuria CKD model. Cilostazol (50 mg/kg/day) or placebo was orally administered once daily from 1-week post-nephrectomy. At 20 weeks, cilostazol significantly attenuated aortic IH as demonstrated by a 34% reduction in the total intima area with 50% and 47% decreases in the ratios of tunica intima area/tunica media area and tunica intima area/(tunica intima + tunica media area), respectively. The diameters of aorta and renal function were unchanged by cilostazol. Interestingly, cilostazol decreased miR-221, but enhanced miR-143 and miR-145 in either in vitro or aortic tissue, as well as attenuated several pro-inflammatory mediators, including asymmetrical dimethylarginine, high-sensitivity C-reactive protein, vascular endothelial growth factor in aorta and serum pro-inflammatory cytokines (IL-6 and TNF-α). We demonstrated a proof of concept of the effectiveness of cilostazol in attenuating IH in a Chr I/R mouse model, a CKD model with predominantly indirect-vascular-injury-induced IH. These considerations warrant further investigation to develop a new primary prevention strategy for CKD-related IH.
Collapse
|
41
|
Afshinnia F, Zeng L, Byun J, Gadegbeku CA, Magnone MC, Whatling C, Valastro B, Kretzler M, Pennathur S. Myeloperoxidase Levels and Its Product 3-Chlorotyrosine Predict Chronic Kidney Disease Severity and Associated Coronary Artery Disease. Am J Nephrol 2017; 46:73-81. [PMID: 28668952 DOI: 10.1159/000477766] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/09/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND The role of myeloperoxidase in chronic kidney disease (CKD) and its association with coronary artery disease (CAD) is controversial. In this study, we compared myeloperoxidase and protein-bound 3-chlorotyrosine (ClY) levels in subjects with varying degrees of CKD and tested their associations with CAD. METHODS From Clinical Phenotyping Resource and Biobank Core, 111 patients were selected from CKD stages 1 to 5. Plasma myeloperoxidase level was measured using enzyme-linked-immunosorbent assay. Plasma protein-bound 3-ClY, a specific product of hypochlorous acid generated by myeloperoxidase was measured by liquid chromatography mass spectrometry. RESULTS We selected 29, 20, 24, 22, and 16 patients from stages 1 to 5 CKD, respectively. In a sex-adjusted general linear model, mean ± SD of myeloperoxidase levels decreased from 18.1 ± 12.3 pmol in stage 1 to 10.9 ± 4.7 pmol in stage 5 (p = 0.011). In patients with and without CAD, the levels were 19.1 ± 10.1 and 14.8 ± 8.7 pmol (p = 0.036). There was an increase in 3-ClY mean from 0.81 ± 0.36 mmol/mol-tyrosine in stage 1 to 1.42 ± 0.41 mmol/mol-tyrosine in stage 5 (p < 0.001). The mean 3-ClY levels in patients with and without CAD were 1.25 ± 0.44 and 1.04 ± 0.42 mmol/mol-tyrosine (p = 0.023), respectively. C-statistic of ClY when added to myeloperoxidase level to predict CKD stage 5 was 0.86, compared to 0.79 for the myeloperoxidase level alone (p = 0.0097). CONCLUSION The myeloperoxidase levels decrease from stages 1 to 5, whereas activity increases. In contrast, both myeloperoxidase and ClY levels rise in the presence of CAD at various stages of CKD. Measuring both plasma myeloperoxidase and 3-CLY levels provide added value to determine the burden of myeloperoxidase-mediated oxidative stress.
Collapse
Affiliation(s)
- Farsad Afshinnia
- University of Michigan, Department of Internal Medicine-Nephrology, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kopprasch S, Bornstein SR, Bergmann S, Graessler J, Hohenstein B, Julius U. Long-term follow-up of circulating oxidative stress markers in patients undergoing lipoprotein apheresis by Direct Adsorption of Lipids (DALI). ATHEROSCLEROSIS SUPP 2017; 30:115-121. [PMID: 29096826 DOI: 10.1016/j.atherosclerosissup.2017.05.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Beyond its well-established efficacy in lowering atherogenic lipids and lipoproteins, DALI (Direct Adsorption of Lipids) apheresis has been shown to have acute anti-inflammatory and endothelium-protective effects. In the present study, we investigated long-term effects of DALI procedures on circulating oxidative stress markers. METHODS Thirteen patients involved in the study underwent regular DALI apheresis for nearly two years. At sessions 1, 40 and 80 conventional lipid status and changes of systemic oxidative stress markers (oxidized LDL, anti-oxidized LDL antibodies, advanced oxidation protein products (AOPP), and myeloperoxidase (MPO)) were examined. RESULTS DALI procedure efficiently reduced atherogenic lipids/lipoproteins. On day three after apheresis lipid parameters returned to pre-apheresis values. They showed no tendency to increase or to decrease over time. No significant differences were found between 1st, 40th and 80th sessions. In a similar way, levels of oxidative stress biomarkers acutely decreased after apheresis sessions and rebounded on day three after apheresis. No significant differences were observed between sessions 1, 40, and 80. CONCLUSION DALI apheresis repeatedly decreases atherogenic lipid/lipoprotein profile and oxidative stress biomarker levels during each session. Among all investigated parameters no longitudinal effects over two years could be observed.
Collapse
Affiliation(s)
- Steffi Kopprasch
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany.
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Sybille Bergmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Juergen Graessler
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Bernd Hohenstein
- Extracorporeal Treatment and Apheresis Center, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Ulrich Julius
- Extracorporeal Treatment and Apheresis Center, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Fetscherstrasse 74, D-01307 Dresden, Germany
| |
Collapse
|
43
|
Abstract
The aim of this chapter is to provide an overview of non-anticoagulant effects of heparins and their potential use in new therapeutic applications. Heparin and heparin derivatives have been tested in inflammatory, pulmonary and reproductive diseases, in cardiovascular, nephro- and neuro-tissue protection and repair, but also as agents against angiogenesis, atheroschlerosis, metastasis, protozoa and viruses. Targeting and inhibition of specific mediators involved in the inflammatory process, promoting some of the above mentioned pathologies, are reported along with recent studies of heparin conjugates and oral delivery systems. Some reports from the institute of the authors, such as those devoted to glycol-split heparins are also included. Among the members and derivatives of this class, several are undergoing clinical trials as antimetastatic and antimalarial agents and for the treatment of labour pain and severe hereditary anaemia. Other heparins, whose therapeutic targets are non-anticoagulant such as nephropathies, retinopathies and cystic fibrosis are also under investigation.
Collapse
Affiliation(s)
| | - Annamaria Naggi
- Istituto di Ricerche Chimiche e Biochimiche G Ronzoni, Milan, Italy.
| |
Collapse
|
44
|
Pregnancy-associated plasma protein-A is a stronger predictor for adverse cardiovascular outcomes after acute coronary syndrome in type-2 diabetes mellitus. Cardiovasc Diabetol 2017; 16:45. [PMID: 28381225 PMCID: PMC5382447 DOI: 10.1186/s12933-017-0526-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/27/2017] [Indexed: 01/05/2023] Open
Abstract
Background The risk prediction of pregnancy-associated plasma protein-A (PAPP-A) for future cardiovascular (CV) events post acute coronary syndrome (ACS) in patients with type-2 diabetes mellitus (T2DM) was investigated in comparison to other risk factors. Methods PAPP-A was measured at hospital admission in 320 consecutive ACS patients (136 with T2DM and 184 without). All patients were followed for 2 years for occurrence of CV death, non-fatal MI or stroke. Effect of PAPP-A on the CV event risk was estimated using Cox regression models. Receiver operating characteristics (ROC) curves were generated to demonstrate the sensitivity and specificity of PAPP-A in predicting CV events. Results ACS patients with T2DM had higher PAPP-A (19.29 ± 16.36 vs. 13.29 ± 13.90 ng/ml, p < 0.001) and higher rate of CV events 2 years post ACS (27.2 vs. 13.6%, p = 0.002) than those without. Higher levels of PAPP-A were significantly associated with increased risk of CV events during 2-year follow-up [HR = 2.97 for 1 SD increase in log10(PAPP-A), 95% CI 2.11–4.18, p < 0.001] in T2DM and (HR = 3.16, 95% CI 2.27–4.39, p < 0.001) in non-T2DM. Among patients with T2DM, PAPP-A showed a larger area under the curve (AUC 0.79) that was significantly more predictive than hsCRP (AUC 0.64), eGFR (AUC 0.66) and LVEF < 50% (AUC 0.52); predictive ability did not improve significantly by including those factors into the model. Conclusions Patients with T2DM had higher levels of PAPP-A and increased risk of CV events. Elevated PAPP-A compared to other risk factors was a stronger predictor for future CV events 2 years post ACS in patients with T2DM. Trial registration ISRCTN10805074. Registered on 20 January 2017, retrospectively registered.
Collapse
|
45
|
Croft DP, Cameron SJ, Morrell CN, Lowenstein CJ, Ling F, Zareba W, Hopke PK, Utell MJ, Thurston SW, Thevenet-Morrison K, Evans KA, Chalupa D, Rich DQ. Associations between ambient wood smoke and other particulate pollutants and biomarkers of systemic inflammation, coagulation and thrombosis in cardiac patients. ENVIRONMENTAL RESEARCH 2017; 154:352-361. [PMID: 28167447 PMCID: PMC5375102 DOI: 10.1016/j.envres.2017.01.027] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 12/25/2016] [Accepted: 01/24/2017] [Indexed: 05/20/2023]
Abstract
BACKGROUND Increased particulate air pollution has been associated with both an increased risk of myocardial infarction (MI) and adverse changes in cardiac biomarkers. Up to 30% of ambient wintertime fine particles (PM2.5) in Rochester, NY are from wood burning. Our study examined associations between ambient levels of a marker of wood smoke (Delta-C) and other particulate air pollutants and biomarkers of inflammation, coagulation and thrombosis. METHODS We measured blood concentrations of C-reactive protein (CRP), D-dimer, fibrinogen, P-selectin, platelet factor 4 (PF-4), von Willebrand factor (vWF), and myeloperoxidase (MPO) of 135 patients undergoing cardiac catheterization during the winters of 2011-2013. We coupled these data with hourly ambient concentrations of Delta-C, black carbon (BC; marker of traffic pollution), and ultrafine (10-100nm; UFP), accumulation mode (100-500nm; AMP), and fine particles (<2.5µm; PM2.5). Using linear regression models, we estimated the change in each biomarker associated with increased pollutant concentrations at intervals between 1 and 96h preceding blood collection. RESULTS Each 0.13µg/m3 increase in Delta-C concentration in the prior 12h was associated with a 0.91% increase in fibrinogen levels (95% CI=0.23%, 1.59%), but unexpectedly in the prior 48h, each 0.17µg/m3 increase in Delta-C concentration was associated with a 2.75% decrease in MPO levels (95% CI=-5.13%,-0.37%). We did not see associations between Delta-C concentrations and any other biomarkers. Interquartile range (IQR) increases in PM2.5, BC, UFP, and AMP concentrations were generally associated with increased CRP and fibrinogen, but not PF4, D-dimer, vWF, or P-selectin. CONCLUSIONS In a population of cardiac patients, we noted adverse changes in fibrinogen associated with increased concentrations of a marker of wood smoke. Increases in PM2.5, BC, AMP, and UFP concentrations in the previous 96h were also associated with adverse changes in markers of systemic inflammation and coagulation, but not with markers of endothelial cell dysfunction or platelet activation.
Collapse
Affiliation(s)
- Daniel P Croft
- Division of Pulmonary and Critical Care, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Scott J Cameron
- Division of Cardiology, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Craig N Morrell
- Division of Cardiology, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Charles J Lowenstein
- Division of Cardiology, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Frederick Ling
- Division of Cardiology, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Wojciech Zareba
- Division of Cardiology, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Philip K Hopke
- Institute for a Sustainable Environment, and Center for Air Resources Engineering and Science, Clarkson University, Box 5708, Potsdam, NY 13699-5708, USA; Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Mark J Utell
- Division of Pulmonary and Critical Care, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Sally W Thurston
- Department of Biostatistics and Computational Biology, University of Rochester, University of Rochester Medical Center, Saunders Research Building, 265 Crittenden Blvd., Box 630, Rochester, NY 14642, USA.
| | - Kelly Thevenet-Morrison
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - Kristin A Evans
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - David Chalupa
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| | - David Q Rich
- Department of Public Health Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA.
| |
Collapse
|
46
|
Maiocchi SL, Morris JC, Rees MD, Thomas SR. Regulation of the nitric oxide oxidase activity of myeloperoxidase by pharmacological agents. Biochem Pharmacol 2017; 135:90-115. [PMID: 28344126 DOI: 10.1016/j.bcp.2017.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/22/2017] [Indexed: 01/10/2023]
Abstract
The leukocyte-derived heme enzyme myeloperoxidase (MPO) is released extracellularly during inflammation and impairs nitric oxide (NO) bioavailability by directly oxidizing NO or producing NO-consuming substrate radicals. Here, structurally diverse pharmacological agents with activities as MPO substrates/inhibitors or antioxidants were screened for their effects on MPO NO oxidase activity in human plasma and physiological model systems containing endogenous MPO substrates/antioxidants (tyrosine, urate, ascorbate). Hydrazide-based irreversible/reversible MPO inhibitors (4-ABAH, isoniazid) or the sickle cell anaemia drug, hydroxyurea, all promoted MPO NO oxidase activity. This involved the capacity of NO to antagonize MPO inhibition by hydrazide-derived radicals and/or the ability of drug-derived radicals to stimulate MPO turnover thereby increasing NO consumption by MPO redox intermediates or NO-consuming radicals. In contrast, the mechanism-based irreversible MPO inhibitor 2-thioxanthine, potently inhibited MPO turnover and NO consumption. Although the phenolics acetaminophen and resveratrol initially increased MPO turnover and NO consumption, they limited the overall extent of NO loss by rapidly depleting H2O2 and promoting the formation of ascorbyl radicals, which inefficiently consume NO. The vitamin E analogue trolox inhibited MPO NO oxidase activity in ascorbate-depleted fluids by scavenging NO-consuming tyrosyl and urate radicals. Tempol and related nitroxides decreased NO consumption in ascorbate-replete fluids by scavenging MPO-derived ascorbyl radicals. Indoles or apocynin yielded marginal effects. Kinetic analyses rationalized differences in drug activities and identified criteria for the improved inhibition of MPO NO oxidase activity. This study reveals that widely used agents have important implications for MPO NO oxidase activity under physiological conditions, highlighting new pharmacological strategies for preserving NO bioavailability during inflammation.
Collapse
Affiliation(s)
- Sophie L Maiocchi
- Mechanisms of Disease & Translational Research, Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jonathan C Morris
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Martin D Rees
- Mechanisms of Disease & Translational Research, Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Shane R Thomas
- Mechanisms of Disease & Translational Research, Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
47
|
Low–Molecular-Weight Heparin for the Prevention of Placenta-mediated Pregnancy Complications. Clin Obstet Gynecol 2017; 60:153-160. [DOI: 10.1097/grf.0000000000000252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Cervantes Gracia K, Llanas-Cornejo D, Husi H. CVD and Oxidative Stress. J Clin Med 2017; 6:E22. [PMID: 28230726 PMCID: PMC5332926 DOI: 10.3390/jcm6020022] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
Nowadays, it is known that oxidative stress plays at least two roles within the cell, the generation of cellular damage and the involvement in several signaling pathways in its balanced normal state. So far, a substantial amount of time and effort has been expended in the search for a clear link between cardiovascular disease (CVD) and the effects of oxidative stress. Here, we present an overview of the different sources and types of reactive oxygen species in CVD, highlight the relationship between CVD and oxidative stress and discuss the most prominent molecules that play an important role in CVD pathophysiology. Details are given regarding common pharmacological treatments used for cardiovascular distress and how some of them are acting upon ROS-related pathways and molecules. Novel therapies, recently proposed ROS biomarkers, as well as future challenges in the field are addressed. It is apparent that the search for a better understanding of how ROS are contributing to the pathophysiology of CVD is far from over, and new approaches and more suitable biomarkers are needed for the latter to be accomplished.
Collapse
Affiliation(s)
- Karla Cervantes Gracia
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Daniel Llanas-Cornejo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
49
|
Abstract
Although substantial improvements have been made in majority of cardiac disorders, heart failure (HF) remains a major health problem, with both increasing incidence and prevalence over the past decades. For that reason, the number of potential biomarkers that could contribute to diagnosis and treatment of HF patients is, almost exponentially, increasing over the recent years. The biomarkers that are, at the moment, more or less ready for use in everyday clinical practice, reflect different pathophysiological processes present in HF. In this review, seven groups of biomarkers associated to myocardial stretch (mid-regional proatrial natriuretic peptide, MR-proANP), myocyte injury (high-sensitive troponins, hs-cTn; heart-type fatty acid-binding protein, H-FABP; glutathione transferase P1, GSTP1), matrix remodeling (galectin-3; soluble isoform of suppression of tumorigenicity 2, sST2), inflammation (growth differentiation factor-15, GDF-15), renal dysfunction (neutrophil gelatinase-associated lipocalin, NGAL; kidney injury molecule-1, KIM-1), neurohumoral activation (adrenomedullin, MR-proADM; copeptin), and oxidative stress (ceruloplasmin; myeloperoxidase, MPO; 8-hydroxy-2'-deoxyguanosine, 8-OHdG; thioredoxin 1, Trx1) in HF will be overviewed. It is important to note that clinical value of individual biomarkers within the single time points in both diagnosis and outcome prediction in HF is limited. Hence, the future of biomarker application in HF lies in the multimarker panel strategy, which would include specific combination of biomarkers that reflect different pathophysiological processes underlying HF.
Collapse
|
50
|
Teng N, Maghzal GJ, Talib J, Rashid I, Lau AK, Stocker R. The roles of myeloperoxidase in coronary artery disease and its potential implication in plaque rupture. Redox Rep 2016; 22:51-73. [PMID: 27884085 PMCID: PMC6837458 DOI: 10.1080/13510002.2016.1256119] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is the main pathophysiological process underlying coronary artery disease (CAD). Acute complications of atherosclerosis, such as myocardial infarction, are caused by the rupture of vulnerable atherosclerotic plaques, which are characterized by thin, highly inflamed, and collagen-poor fibrous caps. Several lines of evidence mechanistically link the heme peroxidase myeloperoxidase (MPO), inflammation as well as acute and chronic manifestations of atherosclerosis. MPO and MPO-derived oxidants have been shown to contribute to the formation of foam cells, endothelial dysfunction and apoptosis, the activation of latent matrix metalloproteinases, and the expression of tissue factor that can promote the development of vulnerable plaque. As such, detection, quantification and imaging of MPO mass and activity have become useful in cardiac risk stratification, both for disease assessment and in the identification of patients at risk of plaque rupture. This review summarizes the current knowledge about the role of MPO in CAD with a focus on its possible roles in plaque rupture and recent advances to quantify and image MPO in plasma and atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathaniel Teng
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia.,b Department of Cardiology , Prince of Wales Hospital , Randwick , New South Wales , Australia
| | - Ghassan J Maghzal
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Jihan Talib
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Imran Rashid
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Antony K Lau
- b Department of Cardiology , Prince of Wales Hospital , Randwick , New South Wales , Australia.,c Faculty of Medicine , University of New South Wales , Sydney , New South Wales , Australia
| | - Roland Stocker
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia.,d School of Medical Sciences , University of New South Wales , Sydney , New South Wales , Australia
| |
Collapse
|