1
|
Sajko S, Skeens E, Schinagl A, Ferhat M, Mirkina I, Mayer J, Rossmueller G, Thiele M, Lisi GP. Redox-dependent plasticity of oxMIF facilitates its interaction with CD74 and therapeutic antibodies. Redox Biol 2024; 75:103264. [PMID: 38972295 PMCID: PMC11263951 DOI: 10.1016/j.redox.2024.103264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024] Open
Abstract
MIF is a ubiquitous protein involved in proinflammatory processes, which undergoes an oxidation-driven conformational change to oxidized (ox)MIF. We demonstrate that hypochlorous acid, produced by neutrophil-released myeloperoxidase (MPO) under inflammatory conditions, effectively oxidizes MIF into the oxMIF isoform, which is specifically recognized by the anti-oxMIF therapeutic antibody, ON104. NMR investigation of MIF oxidized by the MPO system revealed increased flexibility throughout the MIF structure, including at several catalytic and allosteric sites. Mass spectrometry of MPO-oxMIF revealed methionines as the primary site of oxidation, whereas Pro2 and Tyr99/100 remained almost unmodified. ELISA, SPR and cell-based assays demonstrated that structural changes caused by MPO-driven oxidation promoted binding of oxMIF to its receptor, CD74, which does not occur with native MIF. These data reveal the environment and modifications that facilitate interactions between MIF and its pro-inflammatory receptor, and a route for therapeutic intervention targeting the oxMIF isoform.
Collapse
Affiliation(s)
- Sara Sajko
- OncoOne Research and Development GmbH, Vienna, Austria
| | - Erin Skeens
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, USA
| | | | - Maroua Ferhat
- OncoOne Research and Development GmbH, Vienna, Austria
| | - Irina Mirkina
- OncoOne Research and Development GmbH, Vienna, Austria
| | - Julia Mayer
- OncoOne Research and Development GmbH, Vienna, Austria
| | | | | | - George P Lisi
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, USA
| |
Collapse
|
2
|
Wu TJ, Jing X, Teng M, Pritchard KA, Day BW, Naylor S, Teng RJ. Role of Myeloperoxidase, Oxidative Stress, and Inflammation in Bronchopulmonary Dysplasia. Antioxidants (Basel) 2024; 13:889. [PMID: 39199135 PMCID: PMC11351552 DOI: 10.3390/antiox13080889] [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: 07/02/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a lung complication of premature births. The leading causes of BPD are oxidative stress (OS) from oxygen treatment, infection or inflammation, and mechanical ventilation. OS activates alveolar myeloid cells with subsequent myeloperoxidase (MPO)-mediated OS. Premature human neonates lack sufficient antioxidative capacity and are susceptible to OS. Unopposed OS elicits inflammation, endoplasmic reticulum (ER) stress, and cellular senescence, culminating in a BPD phenotype. Poor nutrition, patent ductus arteriosus, and infection further aggravate OS. BPD survivors frequently suffer from reactive airway disease, neurodevelopmental deficits, and inadequate exercise performance and are prone to developing early-onset chronic obstructive pulmonary disease. Rats and mice are commonly used to study BPD, as they are born at the saccular stage, comparable to human neonates at 22-36 weeks of gestation. The alveolar stage in rats and mice starts at the postnatal age of 5 days. Because of their well-established antioxidative capacities, a higher oxygen concentration (hyperoxia, HOX) is required to elicit OS lung damage in rats and mice. Neutrophil infiltration and ER stress occur shortly after HOX, while cellular senescence is seen later. Studies have shown that MPO plays a critical role in the process. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), a reversible MPO inhibitor, attenuates BPD effectively. In contrast, the irreversible MPO inhibitor-AZD4831-failed to provide similar efficacy. Interestingly, KYC cannot offer its effectiveness without the existence of MPO. We review the mechanisms by which this anti-MPO agent attenuates BPD.
Collapse
Affiliation(s)
- Tzong-Jin Wu
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Xigang Jing
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Kirkwood A. Pritchard
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
- Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Billy W. Day
- ReNeuroGen LLC, 2160 San Fernando Dr, Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Dr, Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (T.-J.W.); (X.J.); (M.T.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| |
Collapse
|
3
|
Lessa TLADS, Correia TML, Santos TCD, da Silva RP, Silva BPD, Cavallini MCM, Rocha LS, Souza Peixoto A, Cugnasca BS, Cervi G, Correra TC, Gonçalves AC, Festuccia WTL, Cunha TM, Yatsuda R, de Magalhães ACM, Dos Santos AA, Meotti FC, Queiroz RF. A novel diselenide attenuates the carrageenan-induced inflammation by reducing neutrophil infiltration and the resulting tissue damage in mice. Free Radic Res 2024; 58:229-248. [PMID: 38588405 DOI: 10.1080/10715762.2024.2336566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/06/2024] [Indexed: 04/10/2024]
Abstract
Selenium-containing compounds have emerged as promising treatment for redox-based and inflammatory diseases. This study aimed to investigate the in vitro and in vivo anti-inflammatory activity of a novel diselenide named as dibenzyl[diselanediyIbis(propane-3-1diyl)] dicarbamate (DD). DD reacted with HOCl (k = 9.2 x 107 M-1s-1), like glutathione (k = 1.2 x 108 M-1s-1), yielding seleninic and selenonic acid derivatives, and it also decreased HOCl formation by activated human neutrophils (IC50=4.6 μM) and purified myeloperoxidase (MPO) (IC50=3.8 μM). However, tyrosine, MPO-I and MPO-II substrates, did not restore HOCl formation in presence of DD. DD inhibited the oxidative burst in dHL-60 cells with no toxicity up to 25 µM for 48h. Next, an intraperitoneal administration of 25, 50, and 75 mg/kg DD decreased total leukocyte, neutrophil chemotaxis, and inflammation markers (MPO activity, lipid peroxidation, albumin exudation, nitrite, TNF-α, IL-1β, CXCL1/KC, and CXCL2/MIP-2) on a murine model of carrageenan-induced peritonitis. Likewise, 50 mg/kg DD (i.p.) decreased carrageenan-induced paw edema over 5h. Histological and immunohistochemistry analyses of the paw tissue showed decreased neutrophil count, edema area, and MPO, carbonylated, and nitrated protein staining. Furthermore, DD treatment decreased the fMLP-induced chemotaxis of human neutrophils (IC50=3.7 μM) in vitro with no toxicity. Lastly, DD presented no toxicity in a single-dose model using mice (50 mg/kg, i.p.) over 15 days and in Artemia salina bioassay (50 to 2000 µM), corroborating findings from in silico toxicological study. Altogether, these results demonstrate that DD attenuates carrageenan-induced inflammation mainly by reducing neutrophil migration and the resulting damage from MPO-mediated oxidative burst.
Collapse
Affiliation(s)
- Tássia Liz Araújo Dos Santos Lessa
- Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista, Brazil
| | - Thiago Macêdo Lopes Correia
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Talita Costa Dos Santos
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | | | | | - Maria Cláudia Magalhães Cavallini
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Leonardo Silva Rocha
- Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista, Brazil
| | | | | | - Gustavo Cervi
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Thiago C Correra
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Regiane Yatsuda
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Brazil
- Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | - Amélia Cristina Mendes de Magalhães
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Universidade Federal da Bahia, Vitória da Conquista, Brazil
- Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | | | | | - Raphael Ferreira Queiroz
- Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista, Brazil
- Departamento de Ciências da Saúde, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista, Brazil
| |
Collapse
|
4
|
Jia W, Fu Y, Zhang N, Zhang N, Wang T, Wang Z, Zhang N, Xu J, Yang X, Zhang Q, Li C, Zhang X, Yang W, Han B, Zhang L, Tang N, Bai Z. Ambient PM 2.5-bound polycyclic aromatic hydrocarbons (PAHs) associated with pro-thrombotic biomarkers among young healthy adults: A 16 times repeated measurements panel study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169433. [PMID: 38128672 DOI: 10.1016/j.scitotenv.2023.169433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Studies have shown that the cardio/cerebrovascular toxicity of ambient PM2.5 is related to its bound polycyclic aromatic hydrocarbons (PAHs). Currently, only a few studies have reported the relationship between PM2.5-bound PAHs and promoted blood coagulation and thrombosis, but there isn't a consistent conclusion. Therefore, we conducted a prospective panel study to investigate the association. Thirty-three young healthy adults participated in sixteen repeated visits from 2014 to 2018 in Tianjin, China. During each visit, three pro-thrombotic biomarkers: ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin motif 13), D-dimer and Myeloperoxidase (MPO) were measured. Before each visit, ambient PM2.5 samples were daily collected for one week. Sixteen PAHs were determined using Gas Chromatography-Mass Spectrometer, and the positive matrix factorization (PMF) model was applied to identify the sources. Linear mixed-effects models were fitted to investigate the associations between PM2.5-bound PAHs exposure and the biomarkers. Thirteen time-metrics were defined to identify significant time points of PM2.5-bound PAHs' effects. We observed that the increase of PM2.5-bound PAHs exposure was significantly associated with reduced ADAMTS13, elevated D-dimer and MPO. At lag0, each 5.7 ng/m3 increase in Benzo[j]fluoranthene and per 3.4 ng/m3 increase Dibenz[a,h]anthracene could make a maximum change of -19.08 % in ADAMTS13 and 132.60 % in D-dimer. Additionally, per 16.43 ng/m3 increase in Chrysene could lead to a maximum elevation of 32.14 % in MPO at lag4. The PM2.5-bound PAHs often triggered more significant changes at lag 3,4 and 6. The ambient PM2.5-bound PAHs originated from six sources: coal combustion (43.10 %), biomass combustion (20.77 %), diesel emission (14.78 %), gasoline emission (10.95 %), industrial emission (7.58 %), and cooking emission (2.83 %). The greatest contributors to alterations in ADAMTS13, D-dimer and MPO are industrial emission (-48.43 %), biomass combustion (470.32 %) and diesel emission (13.14 %) at lag4. Our findings indicated that short-term exposure to ambient PM2.5-bound PAHs can induce alterations of pro-thrombotic biomarkers among healthy adults.
Collapse
Affiliation(s)
- Wenhui Jia
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Yucong Fu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Nan Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ningyu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Zhiyu Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Nan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Jia Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Xueli Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Qiang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Changping Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Xumei Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Wen Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Bin Han
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Liwen Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Naijun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Zhipeng Bai
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| |
Collapse
|
5
|
Zheng R, Moynahan K, Georgomanolis T, Pavlenko E, Geissen S, Mizi A, Grimm S, Nemade H, Rehimi R, Bastigkeit J, Lackmann JW, Adam M, Rada-Iglesias A, Nuernberg P, Klinke A, Poepsel S, Baldus S, Papantonis A, Kargapolova Y. Remodeling of the endothelial cell transcriptional program via paracrine and DNA-binding activities of MPO. iScience 2024; 27:108898. [PMID: 38322992 PMCID: PMC10844825 DOI: 10.1016/j.isci.2024.108898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/01/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Myeloperoxidase (MPO) is an enzyme that functions in host defense. MPO is released into the vascular lumen by neutrophils during inflammation and may adhere and subsequently penetrate endothelial cells (ECs) coating vascular walls. We show that MPO enters the nucleus of ECs and binds chromatin independently of its enzymatic activity. MPO drives chromatin decondensation at its binding sites and enhances condensation at neighboring regions. It binds loci relevant for endothelial-to-mesenchymal transition (EndMT) and affects the migratory potential of ECs. Finally, MPO interacts with the RNA-binding factor ILF3 thereby affecting its relative abundance between cytoplasm and nucleus. This interaction leads to change in stability of ILF3-bound transcripts. MPO-knockout mice exhibit reduced number of ECs at scar sites following myocardial infarction, indicating reduced neovascularization. In summary, we describe a non-enzymatic role for MPO in coordinating EndMT and controlling the fate of endothelial cells through direct chromatin binding and association with co-factors.
Collapse
Affiliation(s)
- Ruiyuan Zheng
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Kyle Moynahan
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Theodoros Georgomanolis
- Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Egor Pavlenko
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Simon Geissen
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Athanasia Mizi
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Simon Grimm
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Harshal Nemade
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Rizwan Rehimi
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Jil Bastigkeit
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Jan-Wilm Lackmann
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Cluster of Excellence on Cellular Stress Responses in Age-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Matti Adam
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Alvaro Rada-Iglesias
- Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), University of Cantabria, 39011 Santander, Spain
| | - Peter Nuernberg
- Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, Germany
| | - Anna Klinke
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Simon Poepsel
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Stephan Baldus
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Yulia Kargapolova
- Department III of Internal Medicine, Heart Center, Faculty of Medicine and University Hospital of Cologne, 50937 Cologne, Germany
| |
Collapse
|
6
|
Ortiz-Cerda T, Xie K, Mojadadi A, Witting PK. Myeloperoxidase in Health and Disease. Int J Mol Sci 2023; 24:ijms24097725. [PMID: 37175430 PMCID: PMC10178760 DOI: 10.3390/ijms24097725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Innate and adaptive immune responses comprise a complex network of protein-protein and protein-cell interactions that regulates commensal flora and protects organisms from foreign pathogens and transformed (proliferating) host cells under physiological conditions such as pregnancy, growth and development as well as formulating a response pathological challenge [...].
Collapse
Affiliation(s)
- Tamara Ortiz-Cerda
- Redox Biology Group, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Department of Normal and Pathological Cytology and Histology, Faculty of Medicine, University of Seville, Avenue Sánchez-Pizjuán s/n, 41009 Seville, Spain
| | - Kangzhe Xie
- Redox Biology Group, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Albaraa Mojadadi
- Redox Biology Group, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Paul K Witting
- Redox Biology Group, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Li JX, Tian R, Lu N. Quercetin Attenuates Vascular Endothelial Dysfunction in Atherosclerotic Mice by Inhibiting Myeloperoxidase and NADPH Oxidase Function. Chem Res Toxicol 2023; 36:260-269. [PMID: 36719041 DOI: 10.1021/acs.chemrestox.2c00334] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Myeloperoxidase (MPO) exhibits a unique property to use H2O2 to oxidize chloride and lead to the generation of a strong oxidant, hypochlorous acid (HOCl), which plays important roles in atherosclerosis. A lot of evidence indicates that quercetin, a natural polyphenol derived from human diet, effectively contributes to cardiovascular health. Herein, we found that dietary quercetin significantly inhibited vascular endothelial dysfunction and atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice. Mechanistic studies revealed that dietary quercetin effectively suppressed the MPO level and activity in the vessels of ApoE-/- animals, and p47phox expression and NADPH oxidase activity were simultaneously attenuated after quercetin treatment. In vascular endothelial cells, NADPH oxidase was demonstrated to be the major source of H2O2 formation. Moreover, quercetin effectively attenuated MPO/H2O2-mediated HOCl production and toxicity to human vascular endothelial cells, and this compound was not toxic. The inhibitory effect on MPO activity was likely attributed to that quercetin significantly inhibited NADPH oxidase-derived H2O2 formation in human endothelial cells and could act as an effective mediator for MPO intermediates, subsequently preventing HOCl production by the MPO/H2O2 system. Collectively, it was suggested that quercetin effectively suppressed endothelial dysfunction in atherosclerotic vasculature through the reduction of MPO/NADPH oxidase-mediated HOCl production.
Collapse
Affiliation(s)
- Jia-Xin Li
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| | - Rong Tian
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| | - Naihao Lu
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| |
Collapse
|
8
|
Wang Y, Hammer A, Hoefler G, Malle E, Hawkins CL, Chuang CY, Davies MJ. Hypochlorous Acid and Chloramines Induce Specific Fragmentation and Cross-Linking of the G1-IGD-G2 Domains of Recombinant Human Aggrecan, and Inhibit ADAMTS1 Activity. Antioxidants (Basel) 2023; 12:antiox12020420. [PMID: 36829979 PMCID: PMC9952545 DOI: 10.3390/antiox12020420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease and a leading cause of mortality. It is characterized by arterial wall plaques that contain high levels of cholesterol and other lipids and activated leukocytes covered by a fibrous cap of extracellular matrix (ECM). The ECM undergoes remodelling during atherogenesis, with increased expression of aggrecan, a proteoglycan that binds low-density-lipoproteins (LDL). Aggrecan levels are regulated by proteases, including a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1). Activated leukocytes release myeloperoxidase (MPO) extracellularly, where it binds to proteins and proteoglycans. Aggrecan may therefore mediate colocalization of MPO and LDL. MPO generates hypochlorous acid (HOCl) and chloramines (RNHCl species, from reaction of HOCl with amines on amino acids and proteins) that damage LDL and proteins, but effects on aggrecan have not been examined. The present study demonstrates that HOCl cleaves truncated (G1-IGD-G2) recombinant human aggrecan at specific sites within the IGD domain, with these being different from those induced by ADAMTS1 which also cleaves within this region. Irreversible protein cross-links are also formed dose-dependently. These effects are limited by the HOCl scavenger methionine. Chloramines including those formed on amino acids, proteins, and ECM materials induce similar damage. HOCl and taurine chloramines inactivate ADAMTS1 consistent with a switch from proteolytic to oxidative aggrecan fragmentation. Evidence is also presented for colocalization of aggrecan and HOCl-generated epitopes in advanced human atherosclerotic plaques. Overall, these data show that HOCl and chloramines can induce specific modifications on aggrecan, and that these effects are distinct from those of ADAMTS1.
Collapse
Affiliation(s)
- Yihe Wang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Astrid Hammer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, 8010 Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Clare L. Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Y. Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence: (C.Y.C.); (M.J.D.)
| | - Michael J. Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence: (C.Y.C.); (M.J.D.)
| |
Collapse
|
9
|
Murotomi K, Umeno A, Shichiri M, Tanito M, Yoshida Y. Significance of Singlet Oxygen Molecule in Pathologies. Int J Mol Sci 2023; 24:ijms24032739. [PMID: 36769060 PMCID: PMC9917472 DOI: 10.3390/ijms24032739] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/22/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Reactive oxygen species, including singlet oxygen, play an important role in the onset and progression of disease, as well as in aging. Singlet oxygen can be formed non-enzymatically by chemical, photochemical, and electron transfer reactions, or as a byproduct of endogenous enzymatic reactions in phagocytosis during inflammation. The imbalance of antioxidant enzymes and antioxidant networks with the generation of singlet oxygen increases oxidative stress, resulting in the undesirable oxidation and modification of biomolecules, such as proteins, DNA, and lipids. This review describes the molecular mechanisms of singlet oxygen production in vivo and methods for the evaluation of damage induced by singlet oxygen. The involvement of singlet oxygen in the pathogenesis of skin and eye diseases is also discussed from the biomolecular perspective. We also present our findings on lipid oxidation products derived from singlet oxygen-mediated oxidation in glaucoma, early diabetes patients, and a mouse model of bronchial asthma. Even in these diseases, oxidation products due to singlet oxygen have not been measured clinically. This review discusses their potential as biomarkers for diagnosis. Recent developments in singlet oxygen scavengers such as carotenoids, which can be utilized to prevent the onset and progression of disease, are also described.
Collapse
Affiliation(s)
- Kazutoshi Murotomi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Aya Umeno
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo 693-8501, Japan
| | - Mototada Shichiri
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ikeda 563-8577, Japan
- Correspondence: ; Tel.: +81-72-751-8234
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo 693-8501, Japan
| | | |
Collapse
|
10
|
Porato M, Noël S, Pincemail J, Albert A, Cheramy-Bien JP, Le Goff C, Hamaide A. Selected biomarkers of oxidative stress in healthy Beagle dogs: A preliminary study. Front Vet Sci 2023; 10:1063216. [PMID: 37035819 PMCID: PMC10080027 DOI: 10.3389/fvets.2023.1063216] [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: 10/06/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction While oxidative stress has been studied in pathologic conditions in dogs, data in presumably healthy dogs and standardized protocols are lacking. This work purposed to bridge the gap by presenting provisional physiological ranges for oxidative stress biomarkers in a group of Beagle dogs. Methods Based on our long-standing clinical expertise in the field of oxidative stress, nine plasma biomarkers of oxidative stress were evaluated for their concentrations (mean ± SD) in 14 healthy adult Beagle dogs. Results Selected biomarkers were: vitamins C (7.90 ± 1.36 μg/mL) and E (34.1 ± 6.63 μg/mL), zinc (0.80 ± 0.17 mg/L), copper (0.54 ± 0.048 mg/L), selenium (256 ± 25.7 μg/L), total and oxidized glutathione (822 ± 108 μM and 3.56 ± 1.76 μM), myeloperoxidase (67.4 ± 56.2 ng/mL), and isoprostanes (340 ± 95.3 ng/mL). Glutathione peroxidase activity and superoxide anion production in whole blood were also measured. Glutathione peroxidase activity was 473 ± 34.0 IU/g of hemoglobin and superoxide anion production in whole blood was 18,930 ± 12,742 counts per 30 min. Reduced glutathione/oxidized glutathione and copper/zinc ratios were, respectively, 280 ± 139 and 0.70 ± 0.15. Sex-related differences were recorded for zinc (p = 0.0081), copper/zinc ratio (p = 0.0036) and plasma isoprostanes (p = 0.0045). Conclusion Provisional physiological norms covering 95% of our group were proposed for each biomarker and should be of interest for future studies of canine oxidative stress.
Collapse
Affiliation(s)
- Mathilde Porato
- Clinical Department of Companion Animals, School of Veterinary Medicine, University of Liège, Liège, Belgium
- *Correspondence: Mathilde Porato
| | - Stéphanie Noël
- Clinical Department of Companion Animals, School of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Joël Pincemail
- Departments of Cardiovascular Surgery and Clinical Chemistry, University Hospital, Boulevard de l'Hôpital, Liège, Belgium
| | - Adelin Albert
- Biostatistics, University Hospital, Boulevard de l'Hôpital, Liège, Belgium
| | - Jean-Paul Cheramy-Bien
- Departments of Cardiovascular Surgery and Clinical Chemistry, University Hospital, Boulevard de l'Hôpital, Liège, Belgium
| | - Caroline Le Goff
- Department of Clinical Chemistry, University Hospital, Boulevard de l'Hôpital, Liège, Belgium
| | - Annick Hamaide
- Clinical Department of Companion Animals, School of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
11
|
Xie M, Hao Y, Feng L, Wang T, Yao M, Li H, Ma D, Feng J. Neutrophil Heterogeneity and its Roles in the Inflammatory Network after Ischemic Stroke. Curr Neuropharmacol 2023; 21:621-650. [PMID: 35794770 PMCID: PMC10207908 DOI: 10.2174/1570159x20666220706115957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
As the first peripheral immune cells to enter the brain after ischemic stroke, neutrophils are important participants in stroke-related neuroinflammation. Neutrophils are quickly mobilized from the periphery in response to a stroke episode and cross the blood-brain barrier to reach the ischemic brain parenchyma. This process involves the mobilization and activation of neutrophils from peripheral immune organs (including the bone marrow and spleen), their chemotaxis in the peripheral blood, and their infiltration into the brain parenchyma (including disruption of the blood-brain barrier, inflammatory effects on brain tissue, and interactions with other immune cell types). In the past, it was believed that neutrophils aggravated brain injuries through the massive release of proteases, reactive oxygen species, pro-inflammatory factors, and extracellular structures known as neutrophil extracellular traps (NETs). With the failure of early clinical trials targeting neutrophils and uncovering their underlying heterogeneity, our view of their role in ischemic stroke has become more complex and multifaceted. As neutrophils can be divided into N1 and N2 phenotypes in tumors, neutrophils have also been found to have similar phenotypes after ischemic stroke, and play different roles in the development and prognosis of ischemic stroke. N1 neutrophils are dominant during the acute phase of stroke (within three days) and are responsible for the damage to neural structures via the aforementioned mechanisms. However, the proportion of N2 neutrophils gradually increases in later phases, and this has a beneficial effect through the release of anti-inflammatory factors and other neuroprotective mediators. Moreover, the N1 and N2 phenotypes are highly plastic and can be transformed into each other under certain conditions. The pronounced differences in their function and their high degree of plasticity make these neutrophil subpopulations promising targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Meizhen Xie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Tian Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Mengyue Yao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| |
Collapse
|
12
|
Wang Y, Chuang CY, Hawkins CL, Davies MJ. Activation and Inhibition of Human Matrix Metalloproteinase-9 (MMP9) by HOCl, Myeloperoxidase and Chloramines. Antioxidants (Basel) 2022; 11:antiox11081616. [PMID: 36009335 PMCID: PMC9405048 DOI: 10.3390/antiox11081616] [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] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP9, gelatinase B) plays a key role in the degradation of extracellular-matrix (ECM) proteins in both normal physiology and multiple pathologies, including those linked with inflammation. MMP9 is excreted as an inactive proform (proMMP9) by multiple cells, and particularly neutrophils. The proenzyme undergoes subsequent processing to active forms, either enzymatically (e.g., via plasmin and stromelysin-1/MMP3), or via the oxidation of a cysteine residue in the prodomain (the “cysteine-switch”). Activated leukocytes, including neutrophils, generate O2− and H2O2 and release myeloperoxidase (MPO), which catalyzes hypochlorous acid (HOCl) formation. Here, we examine the reactivity of HOCl and a range of low-molecular-mass and protein chloramines with the pro- and activated forms of MMP9. HOCl and an enzymatic MPO/H2O2/Cl− system were able to generate active MMP9, as determined by fluorescence-activity assays and gel zymography. The inactivation of active MMP9 also occurred at high HOCl concentrations. Low (nM—low μM) concentrations of chloramines formed by the reaction of HOCl with amino acids (taurine, lysine, histidine), serum albumin, ECM proteins (laminin and fibronectin) and basement membrane extracts (but not HEPES chloramines) also activate proMMP9. This activation is diminished by the competitive HOCl-reactive species, methionine. These data indicate that HOCl-mediated oxidation and MMP-mediated ECM degradation are synergistic and interdependent. As previous studies have shown that modified ECM proteins can also stimulate the cellular expression of MMP proteins, these processes may contribute to a vicious cycle of increasing ECM degradation during disease development.
Collapse
Affiliation(s)
- Yihe Wang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
13
|
Xu S, Chuang CY, Malle E, Gamon LF, Hawkins CL, Davies MJ. Influence of plasma halide, pseudohalide and nitrite ions on myeloperoxidase-mediated protein and extracellular matrix damage. Free Radic Biol Med 2022; 188:162-174. [PMID: 35718304 DOI: 10.1016/j.freeradbiomed.2022.06.222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 01/15/2023]
Abstract
Myeloperoxidase (MPO) mediates pathogen destruction by generating the bactericidal oxidant hypochlorous acid (HOCl). Formation of this oxidant is however associated with host tissue damage and disease. MPO also utilizes H2O2 to oxidize other substrates, and we hypothesized that mixtures of other plasma anions, including bromide (Br-), iodide (I-), thiocyanate (SCN-) and nitrite (NO2-), at normal or supplemented concentrations, might modulate MPO-mediated HOCl damage. For the (pseudo)halide anions, only SCN- significantly modulated HOCl formation (IC50 ∼33 μM), which is within the normal physiological range, as judged by damage to human plasma fibronectin or extracellular matrix preparations detected by ELISA and LC-MS. NO2- modulated HOCl-mediated damage, in a dose-dependent manner, at physiologically-attainable anion concentrations. However, this was accompanied by increased tyrosine and tryptophan nitration (detected by ELISA and LC-MS), and the overall extent of damage remained approximately constant. Increasing NO2- concentrations (0.5-20 μM) diminished HOCl-mediated modification of tyrosine and methionine, whereas tryptophan loss was enhanced. At higher NO2- concentrations, enhanced tyrosine and methionine loss was detected. These analytical data were confirmed in studies of cell adhesion and metabolic activity. Together, these data indicate that endogenous plasma levels of SCN- (but not Br- or I-) can modulate protein modification induced by MPO, including the extent of chlorination. In contrast, NO2- alters the type of modification, but does not markedly decrease its extent, with chlorination replaced by nitration. These data also indicate that MPO could be a major source of nitration in vivo, and particularly at inflammatory sites where NO2- levels are often elevated.
Collapse
Affiliation(s)
- Shuqi Xu
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Luke F Gamon
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
14
|
Li P, Zhang C, Wang M, Zhang X, Zhang Y, Tang K, Hu H, Jia X, Zhuang R, Jin B, Ma Y, Zhang Y. Elevation of Myeloperoxidase Correlates with Disease Severity in Patients with Hantaan Virus Infection. Viral Immunol 2022; 35:418-424. [PMID: 35675645 DOI: 10.1089/vim.2022.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hantaan orthohantavirus (HTNV) can cause hemorrhagic fever with renal syndrome (HFRS) characterized by acute kidney injury and hemorrhage. Neutrophils are the most abundant innate immune cell and the body's first line of defense against pathogens. Currently, an increasing number of studies have shown that neutrophils may be a mixed blessing in terms of viral infections. However, the role of neutrophils in HFRS patients with HTNV infection has not been fully declared. In this study, we analyzed plasma levels of both myeloperoxidase (MPO) and MPO-DNA in HFRS patients, together with the clinical parameters. Neutrophil-platelet aggregates (NPAs) during the acute and convalescent phases of HFRS were also assessed. The results showed that plasma MPO-DNA levels had no change in different disease phases or severities of HFRS patients. Whereas plasma MPO significantly increased in the acute phase and critical/severe groups of HFRS patients. Furthermore, plasma MPO was positively correlated with inflammatory clinical parameters, such as white blood cell counts, neutrophil counts, and renal injury-related parameters, such as blood urea nitrogen, blood uric acid, and serum creatinine, as well as negatively correlated with and platelet counts. In addition, NPAs increased both in acute and convalescent phase in HFRS patients compared with normal controls. These results suggested that elevated plasma MPO in HFRS patients correlated with disease severity, together with the increases of NPAs in HFRS patients, which may provide new insights into potential role of neutrophils in the pathogenesis of HFRS.
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China.,Brigade of Cadet, Air Force Medical University, Xi'an, China
| | - Chunmei Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Meng Wang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Xiyue Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Yusi Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Kang Tang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Haifeng Hu
- Center for Infectious Diseases, Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiaozhou Jia
- Department of Infectious Disease, Xi'an Eighth Hospital, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Ying Ma
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| |
Collapse
|
15
|
Yakimov BP, Vlasova II, Efremov YM, Maksimov EG, Shirshin EA, Kagan VE, Timashev PS. Detection of HOCl-driven degradation of the pericardium scaffolds by label-free multiphoton fluorescence lifetime imaging. Sci Rep 2022; 12:10329. [PMID: 35725581 PMCID: PMC9209456 DOI: 10.1038/s41598-022-14138-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/17/2022] [Indexed: 11/24/2022] Open
Abstract
Artificial biomaterials can significantly increase the rate of tissue regeneration. However, implantation of scaffolds leads not only to accelerated tissue healing but also to an immune response of the organism, which results in the degradation of the biomaterial. The synergy of the immune response and scaffold degradation processes largely determines the efficiency of tissue regeneration. Still, methods suitable for fast, accurate and non-invasive characterization of the degradation degree of biomaterial are highly demandable. Here we show the possibility of monitoring the degradation of decellularized bovine pericardium scaffolds under conditions mimicking the immune response and oxidation processes using multiphoton tomography combined with fluorescence lifetime imaging (MPT-FLIM). We found that the fluorescence lifetimes of genipin-induced cross-links in collagen and oxidation products of collagen are prominent markers of oxidative degradation of scaffolds. This was verified in model experiments, where the oxidation was induced with hypochlorous acid or by exposure to activated neutrophils. The fluorescence decay parameters also correlated with the changes of micromechanical properties of the scaffolds as assessed using atomic force microscopy (AFM). Our results suggest that FLIM can be used for quantitative assessments of the properties and degradation of the scaffolds essential for the wound healing processes in vivo.
Collapse
Affiliation(s)
- B P Yakimov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048.,Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskie Gory, Moscow, Russia, 119991
| | - I I Vlasova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048.,Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048
| | - Y M Efremov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048.,Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048
| | - E G Maksimov
- Faculty of Biology, M.V. Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, Russia, 119991
| | - E A Shirshin
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048. .,Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskie Gory, Moscow, Russia, 119991.
| | - V E Kagan
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048.,Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - P S Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048. .,Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8, Moscow, Russia, 119048. .,Faculty of Chemistry, M.V. Lomonosov Moscow State University, 1-3 Leninskie Gory, Moscow, Russia, 119991.
| |
Collapse
|
16
|
Frangie C, Daher J. Role of myeloperoxidase in inflammation and atherosclerosis (Review). Biomed Rep 2022; 16:53. [PMID: 35620311 PMCID: PMC9112398 DOI: 10.3892/br.2022.1536] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Myeloperoxidase (MPO) belongs to the heme peroxidase family, which includes a set of enzymes with potent oxidoreductase activity. MPO is considered an important part of the innate immune system's microbicidal arm and is secreted by neutrophils and macrophages. Interestingly, this enzyme has been implicated in the pathogenesis of several diseases including atherosclerosis. MPO is ubiquitous in atherosclerotic lesions and contributes to the initiation and progression of the disease primarily by oxidizing low-density lipoprotein (LDL) particles. MPO is the only human enzyme with the ability to produce hypochlorous acid (HOCl) at physiological chloride concentrations and HOCl-LDL epitopes were shown to be present inside atheromatous lesions making it a physiologically relevant model for the oxidation of LDL. It has been shown that MPO modified LDL is not able to bind to the native LDL receptor and is recognized instead by scavenger receptors on both endothelial cells and macrophages, which can lead to endothelial dysfunction and foam cell formation, respectively; both of which are instrumental in the progression of the disease. Meanwhile, several studies have proposed MPO as a biomarker for cardiovascular diseases where high levels of this enzyme were linked to an increased risk of developing coronary artery disease. Overall, there is sufficient evidence supporting the value of MPO as a crucial player in health and disease. Thus, future research should be directed towards investigating the still unknown processes associated with this enzyme. This may assist in better understanding the pathophysiological role of MPO, as well in the development of therapeutic strategies for protecting against the deleterious effects of MPO in numerous pathologies such as atherosclerosis.
Collapse
Affiliation(s)
- Christian Frangie
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El‑Koura 100, Lebanon
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El‑Koura 100, Lebanon
| |
Collapse
|
17
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
18
|
Kremserová S, Kocurková A, Chorvátová M, Klinke A, Kubala L. Myeloperoxidase Deficiency Alters the Process of the Regulated Cell Death of Polymorphonuclear Neutrophils. Front Immunol 2022; 13:707085. [PMID: 35211113 PMCID: PMC8860816 DOI: 10.3389/fimmu.2022.707085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 01/18/2022] [Indexed: 01/17/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a key role in host defense. However, their massive accumulation at the site of inflammation can delay regenerative healing processes and can initiate pathological inflammatory processes. Thus, the efficient clearance of PMNs mediated by the induction of regulated cell death is a key process preventing the development of these pathological conditions. Myeloperoxidase (MPO), a highly abundant enzyme in PMN granules, primarily connected with PMN defense machinery, is suggested to play a role in PMN-regulated cell death. However, the contribution of MPO to the mechanisms of PMN cell death remains incompletely characterized. Herein, the process of the cell death of mouse PMNs induced by three different stimuli – phorbol 12-myristate 13-acetate (PMA), opsonized streptococcus (OST), and N-formyl-met-leu-phe (fMLP) – was investigated. MPO-deficient PMNs revealed a significantly decreased rate of cell death characterized by phosphatidylserine surface exposure and cell membrane permeabilization. An inhibitor of MPO activity, 4-aminobenzoic acid hydrazide, did not exhibit a significant effect on PMA-induced cell death compared to MPO deficiency. Interestingly, only the limited activation of markers related to apoptotic cell death was observed (e.g. caspase 8 activation, Bax expression) and they mostly did not correspond to phosphatidylserine surface exposure. Furthermore, a marker characterizing autophagy, cleavage of LC3 protein, as well as histone H3 citrullination and its surface expression was observed. Collectively, the data show the ability of MPO to modulate the life span of PMNs primarily through the potentiation of cell membrane permeabilization and phosphatidylserine surface exposure.
Collapse
Affiliation(s)
- Silvie Kremserová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia
| | - Anna Kocurková
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czechia
| | - Michaela Chorvátová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - 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
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czechia
| |
Collapse
|
19
|
Fatima H, Shahid M, Pruitt C, Pung MA, Mills PJ, Riaz M, Ashraf R. Chemical Fingerprinting, Antioxidant, and Anti-Inflammatory Potential of Hydroethanolic Extract of Trigonella foenum-graecum. Antioxidants (Basel) 2022; 11:364. [PMID: 35204245 PMCID: PMC8869320 DOI: 10.3390/antiox11020364] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/26/2022] [Accepted: 02/07/2022] [Indexed: 02/01/2023] Open
Abstract
In the current study, the antioxidant and anti-inflammatory potential of hydroethanolic extract of T. foenum-graecum seeds was evaluated. Phenolic profiling of T. foenum-graecum was conducted through high-performance liquid chromatography-photodiode array (HPLC-PDA) as well as through the mass spectrometry technique to characterize compounds responsible for bioactivity, which confirmed almost 18 compounds, 13 of which were quantified through a chromatographic assay. In vitro antioxidant analysis of the extract exhibited substantial antioxidant activities with the lowest IC50 value of both DPPH (2,2-diphenyl-1-picrylhydrazyl) and ABTS (2,2'-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid) inhibition assays. The extract was found to be non-toxic against human RBCs and murine macrophage RAW 264.7 cells. Moreover, the extract significantly (p < 0.001) reduced the lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-α), intrlukin-6 (IL-6), prostaglandin E2 (PGE2), and nitric oxide (NO) in RAW 264.7 cells in a concentration-dependent manner. The hydroethanolic extract of T. foenum-graecum exhibited considerable anti-inflammatory potential by decreasing the cellular infiltration to the inflammatory site in both carrageenan-induced peritonitis and an air pouch model of inflammation. Pretreatment with T. foenum-graecum extract caused significant improvement in antioxidants such as superoxide dismutase (SOD), CAT (catalase), malondialdehyde (MDA), and myeloperoxidase (MPO) against oxidative stress induced by carrageenan. Based on our results of in vivo and in vitro experimentation, we concluded that hydroethanolic extract of T. foenum-graecum is a potential source of phenolic compounds with antioxidant and anti-inflammatory potential.
Collapse
Affiliation(s)
- Hina Fatima
- Department of Biochemistry, University of Agriculture, Faisalabad 38000, Pakistan;
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA; (C.P.); (M.A.P.)
- Faculty of Life Sciences, University of Central Punjab, Lahore 54000, Pakistan
| | - Muhammad Shahid
- Department of Biochemistry, University of Agriculture, Faisalabad 38000, Pakistan;
| | - Chris Pruitt
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA; (C.P.); (M.A.P.)
| | - Meredith A. Pung
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA; (C.P.); (M.A.P.)
| | - Paul J. Mills
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA; (C.P.); (M.A.P.)
| | - Muhammad Riaz
- Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha 40100, Pakistan;
| | - Rizwan Ashraf
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan;
| |
Collapse
|
20
|
Song Y, Jiang S, Li C, Loor JJ, Jiang Q, Yang Y, Feng X, Liu S, He J, Wang K, Li Y, Zhang C, Du X, Wang Z, Li X, Liu G. Free fatty acids promote degranulation of azurophil granules in neutrophils by inducing production of NADPH oxidase-derived reactive oxygen species in cows with subclinical ketosis. J Dairy Sci 2022; 105:2473-2486. [PMID: 34998570 DOI: 10.3168/jds.2021-21089] [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: 07/30/2021] [Accepted: 12/06/2021] [Indexed: 11/19/2022]
Abstract
Subclinical ketosis (SCK) in dairy cows, a common metabolic disorder during the peripartal period, is accompanied by systemic inflammation. Excessive release of azurophil granule (AG) contents during degranulation of polymorphonuclear neutrophils (PMN) could contribute to systemic inflammation in SCK cows. Although the increase in blood free fatty acids (FFA) in SCK cows may promote AG degranulation from PMN, the underlying mechanisms are unclear. Thirty multiparous cows (within 3 wk postpartum) with similar lactation numbers (median = 3, range = 2-4) and days in milk (median = 6, range = 3-15) were classified based on serum β-hydroxybutyrate (BHB) level as control (n = 15, BHB < 0.6 mM) or SCK (n = 15, 1.2 mM < BHB < 3.0 mM). Cows with SCK had greater levels of serum haptoglobin, serum amyloid A, IL-1β, IL-6, IL-8 and tumor necrosis factor-α. These proinflammatory factors had strong positive correlations with myeloperoxidase (MPO), a marker protein of PMN AG, whose content was greater in the serum of SCK cows. Both the number of AG and the protein abundance of MPO were lower in PMN isolated from SCK cows. Additionally, we found a greater ratio of blood CH138A+/CD63high cells and greater mean fluorescence intensity of CD63 on the PMN membrane, further confirming the greater degree of AG degranulation in cows with SCK. In vitro FFA dose response (0, 0.3, 0.6, 1.2, and 2.4 mM for 4 h) and time course (0, 0.5, 1, 2, and 4 h with 0.6 mM) experiments were performed on PMN isolated from control cows. The increase in MPO content in extracellular supernatant resulting from those experiments led to the selection of 0.6 mM FFA for 1 h duration as conditions for subsequent studies. After FFA treatment, release of intracellular MPO was increased along with increased levels of CD63 mean fluorescence intensity on the PMN membrane, confirming that FFA promoted degranulation of AG. In addition, FFA treatment increased reactive oxygen species (ROS) production by PMN, an effect that was attenuated by incubation with diphenyleneiodonium chloride (DPI), a NADPH oxidase-derived ROS inhibitor. The mitochondrial-derived ROS inhibitor carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) did not affect ROS in response to FFA treatment. Treatment with FFA increased p47 phosphorylation and mRNA abundance of NCF1, NCF2, and CYBB in PMN. Furthermore, DPI, but not FCCP, dampened the degranulation of PMN AG induced by FFA in vitro. These data suggested that the degranulation of AG in PMN induced by FFA was mediated by NADPH oxidase-derived ROS. As verified ex vivo, PMN from SCK cows had greater levels of ROS, phosphorylation of p47, and mRNA abundance of NCF1, NCF2, and CYBB. Overall, the present study revealed that high blood concentrations of FFA in SCK cows induce the production of NADPH oxidase-derived ROS, thereby promoting degranulation of AG in PMN. The stimulatory effect of FFA on the release of AG content during degranulation, especially MPO, provides a new insight into the systemic inflammation experienced by peripartal cows with SCK.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Shang Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Congyi Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Xiancheng Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Siyuan Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Jiyuan He
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Kexin Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Cai Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003 China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062 China.
| |
Collapse
|
21
|
Pereira JR, da Fonseca AG, de Sena Fernandes LL, Furtado AA, da Silva VC, da Veiga Júnior VF, Sant'Ana AEG, Oliveira CN, Guerra GCB, de Freitas Fernandes-Pedrosa M, Gavioli EC, Oliveira-Costa JF, Soares MBP, de Lima ÁAN, de Melo Silva D, Moura Lemos TMA. Toxicological and pharmacological effects of pentacyclic triterpenes rich fraction obtained from the leaves of Mansoa hirsuta. Biomed Pharmacother 2021; 145:112478. [PMID: 34872801 DOI: 10.1016/j.biopha.2021.112478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/27/2022] Open
Abstract
Mansoa hirsuta is a medicinal plant native to the Brazilian semi-arid region. This approach aimed to investigate the in vitro and in vivo toxicity and anti-inflammatory and analgesic actions of the M. hirsuta fraction (MHF). In vitro cell viability was assessed in 3T3 cells. In vivo, the acute toxicity test, a single dose of the MHF was administered. For the subchronic toxicity test, three doses of were administered for 30 days. Locomotion and motor coordination were assessed using open field and rota-rod. The anti-inflammatory activity was evaluated in carrageenan-induced paw edema and zymosan-induced air-pouch models. Myeloperoxidase (MPO) and total proteins were also measured. The antinociceptive activity MHF was determined using acid acetic-induced abdominal writhing and formalin models. In the cytotoxicity assay, MHF showed no significative impairment of cell viability and in the acute toxicity study, did not cause mortality or signs of toxicity. Repeated exposure to MHF did not cause relevant toxicological changes. The evaluation in the open field test showed that the MHF did not alter the locomotor activity and there was no change in motor coordination and balance of animals. MHF significantly reduced edema, MPO production, the migration of leukocytes and protein leakage. In addition, MHF reduced abdominal writhing and significantly inhibited the first and second stage of the formalin test. The results of this study indicated that MHF has an anti-inflammatory and analgesic potential without causing acute or subchronic toxic effects and it can be a promising natural source to be explored.
Collapse
Affiliation(s)
- Joquebede Rodrigues Pereira
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Aldilane Gonçalves da Fonseca
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Luzia Leiros de Sena Fernandes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Allanny Alves Furtado
- Department of Pharmacy, Laboratório Escola de Farmácia Industrial, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Valéria Costa da Silva
- Department of Biophysics and Pharmacology, Centro de Biociências, Federal University of Rio Grande do Norte, Natal, RN 59078-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Valdir Florêncio da Veiga Júnior
- Chemical Engineering Department, Military Institute of Engineering, Rio de Janeiro, RJ 22290-270, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Antônio Euzébio Goulart Sant'Ana
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus A.C. Simões, Maceió, AL 57072-970, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Cláudia Nunes Oliveira
- Department of Pathology, Federal University of Rio Grande do Norte, Natal, RN 59012570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Gerlane Coelho Bernardo Guerra
- Department of Biophysics and Pharmacology, Centro de Biociências, Federal University of Rio Grande do Norte, Natal, RN 59078-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Matheus de Freitas Fernandes-Pedrosa
- Department of Pharmacy, Laboratório Escola de Farmácia Industrial, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Centro de Biociências, Federal University of Rio Grande do Norte, Natal, RN 59078-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil
| | - José Fernando Oliveira-Costa
- State Health Secretary, Pharmaceutical Assistance Directory, Salvador, Bahia 41233-015, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Milena Botelho Pereira Soares
- State Health Secretary, Pharmaceutical Assistance Directory, Salvador, Bahia 41233-015, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Ádley Antonini Neves de Lima
- Department of Pharmacy, Laboratório Escola de Farmácia Industrial, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Daniel de Melo Silva
- Gonçalo Moniz Center, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia 40296-710, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| | - Telma Maria Araújo Moura Lemos
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Norte, Natal, RN 59012-570, Brazil; Department of Chemistry and Exact, State University of Southwest of Bahia, Jequié, BA 45208-091, Brazil.
| |
Collapse
|
22
|
Inflammatory Mediators of Platelet Activation: Focus on Atherosclerosis and COVID-19. Int J Mol Sci 2021; 22:ijms222011170. [PMID: 34681830 PMCID: PMC8539848 DOI: 10.3390/ijms222011170] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Atherosclerotic cardiovascular diseases are characterized by a dysregulated inflammatory and thrombotic state, leading to devastating complications with increased morbidity and mortality rates. Summary: In this review article, we present the available evidence regarding the impact of inflammation on platelet activation in atherosclerosis. Key messages: In the context of a dysfunctional vascular endothelium, structural alterations by means of endothelial glycocalyx thinning or functional modifications through impaired NO bioavailability and increased levels of von Willebrand factor result in platelet activation. Moreover, neutrophil-derived mediators, as well as neutrophil extracellular traps formation, have been implicated in the process of platelet activation and platelet-leukocyte aggregation. The role of pro-inflammatory cytokines is also critical since their receptors are also situated in platelets while TNF-α has also been found to induce inflammatory, metabolic, and bone marrow changes. Additionally, important progress has been made towards novel concepts of the interaction between inflammation and platelet activation, such as the toll-like receptors, myeloperoxidase, and platelet factor-4. The accumulating evidence is especially important in the era of the coronavirus disease-19 pandemic, characterized by an excessive inflammatory burden leading to thrombotic complications, partially mediated by platelet activation. Lastly, recent advances in anti-inflammatory therapies point towards an anti-thrombotic effect secondary to diminished platelet activation.
Collapse
|
23
|
Kameda T, Horiuchi Y, Shimano S, Yano K, Lai SJ, Ichimura N, Tohda S, Kurihara Y, Tozuka M, Ohkawa R. Effect of myeloperoxidase oxidation and N-homocysteinylation of high-density lipoprotein on endothelial repair function. Biol Chem 2021; 403:265-277. [PMID: 34448387 DOI: 10.1515/hsz-2021-0247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Endothelial cell (EC) migration is essential for healing vascular injuries. Previous studies suggest that high-density lipoprotein (HDL) and apolipoprotein A-I (apoA-I), the major protein constituent of HDL, have endothelial healing functions. In cardiovascular disease, HDL is modified by myeloperoxidase (MPO) and N-homocysteine, resulting in apoA-I/apoA-II heterodimer and N-homocysteinylated (N-Hcy) apoA-I formation. This study investigated whether these modifications attenuate HDL-mediated endothelial healing. Wound healing assays were performed to analyze the effect of MPO-oxidized HDL and N-Hcy HDL in vitro. HDL obtained from patients with varying troponin I levels were also examined. MPO-oxidized HDL reduces EC migration compared to normal HDL in vitro, and N-Hcy HDL showed a decreasing trend toward EC migration. EC migration after treatment with HDL from patients was decreased compared to HDL isolated from healthy controls. Increased apoA-I/apoA-II heterodimer and N-Hcy apoA-I levels were also detected in HDL from patients. Wound healing cell migration was significantly negatively correlated with the ratio of apoA-I/apoA-II heterodimer to total apoA-II and N-Hcy apoA-I to total apoA-I. MPO-oxidized HDL containing apoA-I/apoA-II heterodimers had a weaker endothelial healing function than did normal HDL. These results indicate that MPO-oxidized HDL and N-Hcy HDL play a key role in the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Takahiro Kameda
- Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yuna Horiuchi
- Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Department of Clinical Laboratory Medicine, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu City, Chiba, 279-0021, Japan
| | - Shitsuko Shimano
- Clinical Laboratory, Medical Hospital, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kouji Yano
- Division of Clinical Medicine, Research and Education Center for Clinical Pharmacy, Kitasato University School of Pharmacy, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Shao-Jui Lai
- Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Naoya Ichimura
- Clinical Laboratory, Medical Hospital, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shuji Tohda
- Clinical Laboratory, Medical Hospital, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Yuriko Kurihara
- Department of Medical Technology, School of Health Sciences, Tokyo University of Technology, 5-23-22 Nishikamata, Ota-ku, Tokyo, 144-8535, Japan
| | - Minoru Tozuka
- Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Life Science Research Center, Nagano Children's Hospital, 3100 Toyoshina, Azumino, 399-8288, Japan
| | - Ryunosuke Ohkawa
- Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| |
Collapse
|
24
|
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: 56] [Impact Index Per Article: 14.0] [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
|
25
|
Thioredoxin-1 and Correlations of the Plasma Cytokines Regarding Aortic Valve Stenosis Severity. Biomedicines 2021; 9:biomedicines9081041. [PMID: 34440245 PMCID: PMC8391645 DOI: 10.3390/biomedicines9081041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022] Open
Abstract
Aortic valve stenosis (AS) develops not only with a pronounced local inflammatory response, but also oxidative stress is involved. The aim of this study was to evaluate the plasma levels of thioredoxin-1 (TRX1), myeloperoxidase (MPO), chemerin, growth differentiation factor 15 (GDF-15), angiopoietin-2 (Ang-2), vascular endothelial growth factor A (VEGF-A), fibroblast growth factor 2 (FGF-2), fibroblast growth factor 21 (FGF-21), and metalloproteinase (MMP)-1, -3, and -9 in acquired AS patients as well as to clarify the correlations of TXR1 and the plasma inflammatory biomarkers regarding AS severity. AS patients were classified into three groups: 16 patients with mild AS stenosis, 19 with moderate and 11 with severe AS, and 30 subjects without AS were selected as a control group. AS patients had significantly higher plasma levels of TRX1 compared to controls, but the highest difference was found in mild AS patients compared to the controls. We conclude that AS is associated with significantly increased plasma TRX1 levels, and TRX1 might serve as a specific and sensitive biomarker of AS. TRX1 and also chemerin, GDF-15, VEGF-A, FGF-2 and FGF-21 significantly correlate with AS severity degrees. TRX1 also showed positive association with FGF-2, VEGF-A, and MMP-3 in all AS patients.
Collapse
|
26
|
Peng L, Li X, Li Y, Zhao W, Nie S, Yu H, Qi Y, Qin Y, Zhang H. Increased concentrations of myeloperoxidase in serum and serum extracellular vesicles are associated with type 2 diabetes mellitus. Clin Chim Acta 2021; 522:70-76. [PMID: 34390687 DOI: 10.1016/j.cca.2021.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/31/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inflammatory response plays a critical role in the initiation and progression of type 2 diabetes mellitus (T2DM). Myeloperoxidase (MPO), a leukocyte-derived protagonist, exerts its proinflammatory properties in many complications. We explored the associations between serum extracellular vesicle (EV)-derived MPO as well as serum MPO and T2DM. METHODS We performed a cross-sectional study in 151 individuals, including 93 patients with T2DM and 58 non-T2DM controls. The concentrations of serum EV-derived MPO and serum MPO were measured by Luminex Assay. RESULTS Our data showed that serum EV-derived MPO concentrations and serum MPO concentrations were significantly higher in T2DM patients compared with non-T2DM subjects. In addition, multivariate logistic regression analysis revealed that serum EV-derived MPO as well as serum MPO was independently associated with the presence of T2DM even after adjusting for confounding factors (OR = 1.836 /1 ng EV-derived MPO, 95% CI = 1.395-2.417, P < 0.001; OR = 4.135 /10 ng serum MPO, 95% CI = 2.285-7.483, P < 0.001). Furthermore, serum MPO showed marginally higher discriminatory accuracy than serum EV-derived MPO in screening T2DM (AUC = 0.858; AUC = 0.779). CONCLUSION Increased concentrations of the inflammatory marker MPO either in serum or in serum EVs were independently associated with T2DM.
Collapse
Affiliation(s)
- Lu Peng
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xinwei Li
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wen Zhao
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shaoping Nie
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, China
| | - Huahui Yu
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yue Qi
- Department of Epidemiology, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Huina Zhang
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Trentini A, Rosta V, Spadaro S, Bellini T, Rizzo P, Vieceli Dalla Sega F, Passaro A, Zuliani G, Gentili V, Campo G, Cervellati C. Development, optimization and validation of an absolute specific assay for active myeloperoxidase (MPO) and its application in a clinical context: role of MPO specific activity in coronary artery disease. Clin Chem Lab Med 2021; 58:1749-1758. [PMID: 32031967 DOI: 10.1515/cclm-2019-0817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/04/2020] [Indexed: 12/13/2022]
Abstract
Background Myeloperoxidase (MPO) is an enzyme with a recognized prognostic role in coronary artery disease (CAD), which is also emerging as a promising biomarker for cardiac risk stratification. However, the lack of a consensus method for its quantification has hindered its implementation in clinical practice. The aim of our work was to optimize an absolute sensitive assay for active MPO without external standards, to validate the method in the clinical context of CAD patients, and to estimate the enzyme specific activity. Methods In order to determine the MPO concentration using fluorescence readings, this ELISA assay exploits the activity of the enzyme recognized by specific antibodies. The assay was validated in a small cohort of patients that included: healthy subjects (n=60); patients with acute myocardial infarction (AMI, n=25); patients with stable CAD (SCAD, n=25) and a concomitant chronic obstructive pulmonary disease (COPD). Then, total MPO concentration and specific activity (activity/total MPO) were determined. Results The assay showed an intra- and inter-assay coefficient of variation of 5.8% and 10.4%, respectively, with a limit of detection (LoD) of 0.074 μU. Both AMI and SCAD patients had higher active and total MPO than controls (p<0.0001 and p<0.01, respectively). The specific activity of MPO was higher in SCAD patients compared to both controls and AMI (p<0.0001). Conclusions The study presents a robust and sensitive method for assaying MPO activity in biological fluids with low variability. Moreover, the determination of the specific activity could provide novel insight into the role of MPO in cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy
| | - Valentina Rosta
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy
| | - Savino Spadaro
- Department of Morphology, Surgery and Experimental Medicine, Section of Anesthesia and Intensive Care University of Ferrara, Ferrara, Italy
| | - Tiziana Bellini
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | | | - Angelina Passaro
- Department of Medical Sciences, Section of Internal and Cardio Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Valentina Gentili
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy.,Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy
| |
Collapse
|
28
|
Lycopene suppresses palmitic acid-induced brain oxidative stress, hyperactivity of some neuro-signalling enzymes, and inflammation in female Wistar rat. Sci Rep 2021; 11:15038. [PMID: 34294819 PMCID: PMC8298469 DOI: 10.1038/s41598-021-94518-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/29/2021] [Indexed: 01/14/2023] Open
Abstract
Neuroinflammation can be triggered by certain high caloric nutrients such as palmitic acid (PA). The effect of lycopene against PA-induced neuroinflammation in female rats has not been as explored. In the present study, thirty rats (weighing 150–200) g were randomly allotted into six groups (n = 5) comprising normal control, PA control, PA + lycopene (0.24 mg/kg), PA + lycopene (0.48 mg/kg), lycopene (0.24 mg/kg), and lycopene (0.48 mg/kg), respectively. After seven weeks of PA challenge (5 mM) including two weeks of lycopene treatment, the brain was excised for analyses. Palmitic acid overload caused significant (p < 0.05) increases in adenosine deaminase, monoamine oxidase-A, nucleotides tri-phosphatase, 5′-nucleotidase, acetylcholine esterase, and myeloperoxidase activities, and malondialdehyde (MDA) levels which were reduced significantly in the lycopene-treated groups. Conversely, catalase and glutathione peroxidase activities, and reduced glutathione levels concentration decreased by 43%, 34%, and 12%, respectively in the PA control groups compared with the Control. Also, PA triggered a decrease in the brain phospholipids (11.43%) and cholesterol (11.11%), but increased triacylglycerol level (50%). Furthermore, upregulated expressions of Interleukin-1β, Interleukin-6, and NF-ĸB-p65 in the PA control were attenuated, while decreased Interleukine-10 expression was upregulated due to lycopene treatment. Severe brain vacuolation observed in the histology of the PA control rats was normalized by lycopene. This study concludes that lycopene ameliorated PA-induced neuroinflammation, probably via attenuation of oxidative stress, and downregulation of TLR4/ NF-κB -p65 axis.
Collapse
|
29
|
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
|
30
|
Morales-Primo AU, Becker I, Zamora-Chimal J. Neutrophil extracellular trap-associated molecules: a review on their immunophysiological and inflammatory roles. Int Rev Immunol 2021; 41:253-274. [PMID: 34036897 DOI: 10.1080/08830185.2021.1921174] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophil extracellular traps (NETs) are a defense mechanism against pathogens. They are composed of DNA and various proteins and have the ability to hinder microbial spreading and survival. However, NETs are not only related to infections but also participate in sterile inflammatory events. In addition to DNA, NETs contain histones, serine proteases, cytoskeletal proteins and antimicrobial peptides, all of which have immunomodulatory properties that can augment or decrease the inflammatory response. Extracellular localization of these molecules alerts the immune system of cellular damage, which is triggered by recognition of damage-associated molecular patterns (DAMPs) through specific pattern recognition receptors. However, not all of these molecules are DAMPs and may have other immunophysiological properties in the extracellular space. The release of NETs can lead to production of pro-inflammatory cytokines (due to TLR2/4/9 and inflammasome activation), the destruction of the extracellular matrix, activation of serine proteases and of matrix metallopeptidases (MMPs), modulation of cellular proliferation, induction of cellular migration and adhesion, promotion of thrombogenesis and angiogenesis and disruption of epithelial and endothelial permeability. Understanding the dynamics of NET-associated molecules, either individually or synergically, will help to unravel their role in inflammatory events and open novel perspectives for potential therapeutic targets. We here review molecules contained within NETS and their immunophysiological roles.
Collapse
Affiliation(s)
- Abraham U Morales-Primo
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| | - Ingeborg Becker
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| |
Collapse
|
31
|
Jin Z, Zhou L, Tian R, Lu N. Myeloperoxidase Targets Apolipoprotein A-I for Site-Specific Tyrosine Chlorination in Atherosclerotic Lesions and Generates Dysfunctional High-Density Lipoprotein. Chem Res Toxicol 2021; 34:1672-1680. [PMID: 33861588 DOI: 10.1021/acs.chemrestox.1c00086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We previously demonstrated that apolipoprotein A-I (apoA-I), the major protein component of high-density lipoprotein (HDL), is an important target for myeloperoxidase (MPO)-catalyzed tyrosine chlorination in the circulation of subjects with cardiovascular diseases. Oxidation of apoA-I by MPO has been reported to deprive HDL of its protective properties. However, the potential effects of MPO-mediated site-specific tyrosine chlorination of apoA-I on dysfunctional HDL formation and atherosclerosis was unclear. Herein, Tyr192 in apoA-I was found to be the major chlorination site in both lesion and plasma HDL from humans with atherosclerosis, while MPO binding to apoA-I was demonstrated by immunoprecipitation studies in vivo. In vitro, MPO-mediated damage of lipid-free apoA-I impaired its ability to promote cellular cholesterol efflux by the ABCA1 pathway, whereas oxidation to lipid-associated apoA-I inhibited lecithin:cholesterol acyltransferase activation, two key steps in reverse cholesterol transport. Compared with native apoA-I, apoA-I containing a Tyr192 → Phe mutation was moderately resistant to oxidative inactivation by MPO. In high-fat-diet-fed apolipoprotein E-deficient mice, compared with native apoA-I, subcutaneous injection with oxidized apoA-I (MPO treated) failed to mediate the lipid content in aortic plaques while mutant apoA-I (Tyr192 → Phe) showed a slightly stronger ability to reduce the lipid content in vivo. Our observations suggest that oxidative damage of apoA-I and HDL involves MPO-dependent site-specific tyrosine chlorination, raising the feasibility of producing MPO-resistant forms of apoA-I that have stronger antiatherosclerotic activity in vivo.
Collapse
Affiliation(s)
- Zelong Jin
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China
| | - Lan Zhou
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China
| | - Rong Tian
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China
| | - Naihao Lu
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China
| |
Collapse
|
32
|
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
|
33
|
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: 7] [Impact Index Per Article: 1.8] [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
|
34
|
Santos MBD, Carvalho Marques B, Miranda Ayusso G, Rocha Garcia MA, Chiquetto Paracatu L, Pauli I, Silva Bolzani V, Defini Andricopulo A, Farias Ximenes V, Zeraik ML, Regasini LO. Chalcones and their B-aryl analogues as myeloperoxidase inhibitors: In silico, in vitro and ex vivo investigations. Bioorg Chem 2021; 110:104773. [PMID: 33744807 DOI: 10.1016/j.bioorg.2021.104773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 01/29/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022]
Abstract
In the present study, a series of chalcones and their B-aryl analogues were prepared and evaluate as inhibitors of myeloperoxidase (MPO) chlorinating activity, using in vitro and ex vivo assays. Among these, B-thiophenyl chalcone (analogue 9) demonstrated inhibition of in vitro and ex vivo MPO chlorinating activity, exhibiting IC50 value of 0.53 and 19.2 µM, respectively. Potent ex vivo MPO inhibitors 5, 8 and 9 were not toxic to human neutrophils at 50 µM, as well as displayed weak 2,2-diphenyl-1-pycrylhydrazyl radical (DPPH•) and hypochlorous acid (HOCl) scavenger abilities. Docking simulations indicated binding mode of MPO inhibitors, evidencing hydrogen bonds between the amino group at 4'position (ring A) of chalcones with Gln91, Asp94, and Hys95 MPO residues. In this regard, the efficacy and low toxicity promoted aminochalcones and arylic analogues to the rank of hit compounds in the search for new non-steroidal anti-inflammatory compounds.
Collapse
Affiliation(s)
- Mariana Bastos Dos Santos
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), 15054-000 São José do Rio Preto, SP, Brazil
| | - Beatriz Carvalho Marques
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), 15054-000 São José do Rio Preto, SP, Brazil
| | - Gabriela Miranda Ayusso
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), 15054-000 São José do Rio Preto, SP, Brazil
| | - Mayara Aparecida Rocha Garcia
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), 15054-000 São José do Rio Preto, SP, Brazil
| | - Luana Chiquetto Paracatu
- Department of Chemistry, Faculty of Sciences, São Paulo State University (UNESP), 17033-360 Bauru, SP, Brazil
| | - Ivani Pauli
- Physics Institute of São Carlos, University of São Paulo, 13563-120 São Carlos, SP, Brazil
| | - Vanderlan Silva Bolzani
- Department of Organic Chemistry, Institute of Chemistry, São Paulo State University, 14800-900 Araraquara, SP, Brazil
| | | | - Valdecir Farias Ximenes
- Department of Chemistry, Faculty of Sciences, São Paulo State University (UNESP), 17033-360 Bauru, SP, Brazil
| | - Maria Luiza Zeraik
- Department of Chemistry, State University of Londrina (UEL), 86051-990 Londrina, PR, Brazil.
| | - Luis Octavio Regasini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), 15054-000 São José do Rio Preto, SP, Brazil.
| |
Collapse
|
35
|
Flouda K, Gammelgaard B, Davies MJ, Hawkins CL. Modulation of hypochlorous acid (HOCl) induced damage to vascular smooth muscle cells by thiocyanate and selenium analogues. Redox Biol 2021; 41:101873. [PMID: 33550113 PMCID: PMC7868818 DOI: 10.1016/j.redox.2021.101873] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
The production of hypochlorous acid (HOCl) by myeloperoxidase (MPO) plays a key role in immune defense, but also induces host tissue damage, particularly in chronic inflammatory pathologies, including atherosclerosis. This has sparked interest in the development of therapeutic approaches that decrease HOCl formation during chronic inflammation, including the use of alternative MPO substrates. Thiocyanate (SCN−) supplementation decreases HOCl production by favouring formation of hypothiocyanous acid (HOSCN), which is more selectively toxic to bacterial cells. Selenium-containing compounds are also attractive therapeutic agents as they react rapidly with HOCl and can be catalytically recycled. In this study, we examined the ability of SCN−, selenocyanate (SeCN−) and selenomethionine (SeMet) to modulate HOCl-induced damage to human coronary artery smooth muscle cells (HCASMC), which are critical to both normal vessel function and lesion formation in atherosclerosis. Addition of SCN− prevented HOCl-induced cell death, altered the pattern and extent of intracellular thiol oxidation, and decreased perturbations to calcium homeostasis and pro-inflammatory signaling. Protection was also observed with SeCN− and SeMet, though SeMet was less effective than SeCN− and SCN−. Amelioration of damage was detected with sub-stoichiometric ratios of the added compound to HOCl. The effects of SCN− are consistent with conversion of HOCl to HOSCN. Whilst SeCN− prevented HOCl-induced damage to a similar extent to SCN−, the resulting product hyposelenocyanous acid (HOSeCN), was more toxic to HCASMC than HOSCN. These results provide support for the use of SCN− and/or selenium analogues as scavengers, to decrease HOCl-induced cellular damage and HOCl production at inflammatory sites in atherosclerosis and other pathologies. HOCl induces extensive smooth muscle cell death and irreversible thiol oxidation. Addition of SCN− decreases the extent of HOCl-induced cell damage. SeCN− has similar protective effects to SCN− towards HOCl-induced cell damage. HOSeCN is less toxic than HOCl but more damaging than HOSCN. SeMet modulates HOCl-induced damage but less effectively than SCN− or SeCN−.
Collapse
Affiliation(s)
- Konstantina Flouda
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark
| | - Bente Gammelgaard
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, DK-2200, Denmark.
| |
Collapse
|
36
|
Tian R, Jin Z, Zhou L, Zeng XP, Lu N. Quercetin Attenuated Myeloperoxidase-Dependent HOCl Generation and Endothelial Dysfunction in Diabetic Vasculature. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:404-413. [PMID: 33395297 DOI: 10.1021/acs.jafc.0c06335] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Myeloperoxidase (MPO)-dependent hypochlorous acid (HOCl) generation plays crucial roles in diabetic vascular complications. As a natural polyphenol, quercetin has antioxidant properties in various diabetic models. Herein, we investigated the therapeutic mechanism for quercetin on MPO-mediated HOCl generation and endothelial dysfunction in diabetic vasculature. In vitro, the presence of MPO could amplify high glucose-induced endothelial dysfunction which was significantly inhibited by the NADPH oxidase inhibitor, HOCl or H2O2 scavengers, revealing the contribution of MPO/H2O2/HOCl to vascular endothelial injury. Furthermore, quercetin effectively inhibited MPO/high glucose-mediated HOCl generation and cytotoxicity to vascular endothelial cells. The inhibitive effect on MPO activity was related to the fact that quercetin reduced high glucose-induced H2O2 generation in endothelial cells and directly acted as a competitive substrate for MPO, thus limiting MPO/H2O2-dependent HOCl production. Moreover, quercetin could attenuate HOCl-caused endothelial dysfunction in endothelial cells and isolated aortas. In vivo, dietary quercetin significantly inhibited aortic endothelial dysfunction in diabetic mice, while this compound simultaneously suppressed vascular MPO expression and activity. Therefore, it was demonstrated herein that quercetin inhibited endothelial injury in diabetic vasculature via suppression of MPO/high glucose-dependent HOCl formation.
Collapse
Affiliation(s)
- Rong Tian
- MOE Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Zeran Jin
- MOE Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Lan Zhou
- MOE Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Xing-Ping Zeng
- MOE Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| | - Naihao Lu
- MOE Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| |
Collapse
|
37
|
Finelli R, Darbandi S, Pushparaj PN, Henkel R, Ko E, Agarwal A. In Silico Sperm Proteome Analysis to Investigate DNA Repair Mechanisms in Varicocele Patients. Front Endocrinol (Lausanne) 2021; 12:757592. [PMID: 34975746 PMCID: PMC8719329 DOI: 10.3389/fendo.2021.757592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/30/2021] [Indexed: 01/19/2023] Open
Abstract
Varicocele, a condition associated with increased oxidative stress, negatively affects sperm DNA integrity and reduces pregnancy rates. However, the molecular mechanisms related to DNA integrity, damage, and repair in varicocele patients remain unclear. This study aimed to determine the role of DNA repair molecular mechanisms in varicocele-related infertility by combining an in silico proteomics approach with wet-laboratory techniques. Proteomics results previously generated from varicocele patients (n=50) and fertile controls (n=10) attending our Andrology Center were reanalyzed using bioinformatics tools, including the WEB-based Gene SeT AnaLysis Toolkit, Open Target Platform, and Ingenuity Pathway Analysis (IPA), to identify differentially expressed proteins (DEPs) involved in DNA repair. Subsequently, selected DEPs in spermatozoa were validated using western blotting in varicocele (n = 13) and fertile control (n = 5) samples. We identified 99 DEPs mainly involved in male reproductive system disease (n=66) and male infertility (n=47). IPA analysis identified five proteins [fatty acid synthase (FASN), myeloperoxidase (MPO), mitochondrial aconitate hydratase (ACO2), nucleoporin 93 (NUP93), and 26S proteasome non-ATPase regulatory subunit 14 (PSMD14)] associated with DNA repair deficiency, which showed altered expression in varicocele (P <0.03). We validated ACO2 downregulation (fold change=0.37, change%=-62.7%, P=0.0001) and FASN overexpression (fold change = 4.04, change %= 303.7%, P = 0.014) in men with varicocele compared to controls. This study combined a unique in silico approach with an in vitro validation of the molecular mechanisms that may be responsible for varicocele-associated infertility. We identified ACO2 and FASN as possible proteins involved in DNA repair, whose altered expression may contribute to DNA damage in varicocele pathophysiology.
Collapse
Affiliation(s)
- Renata Finelli
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Sara Darbandi
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran
- Gene Therapy and Regenerative Medicine Research Center, Hope Generation Foundation, Tehran, Iran
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research and Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ralf Henkel
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, United States
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
- LogixX Pharma, Reading, United Kingdom
| | - Edmund Ko
- Department of Urology, Loma Linda University Health, Loma Linda, CA, United States
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, United States
- *Correspondence: Ashok Agarwal,
| |
Collapse
|
38
|
Kumar VP, Stone S, Biswas S, Sharma N, Ghosh SP. Gamma Tocotrienol Protects Mice From Targeted Thoracic Radiation Injury. Front Pharmacol 2020; 11:587970. [PMID: 33343356 PMCID: PMC7748112 DOI: 10.3389/fphar.2020.587970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/06/2020] [Indexed: 01/20/2023] Open
Abstract
Radiation injury will result in multiorgan dysfuntion leading to multiorgan failure. In addition to many factors such as radiation dose, dose rate, the severity of the injury will also depend on organ systems which are exposed. Here, we report the protective property of gamma tocotrienol (GT3) in total as well as partial body irradiation (PBI) model in C3H/HeN male mice. We have carried out PBI by targeting thoracic region (lung-PBI) using Small Animal Radiation Research Platform, an X-ray irradiator with capabilities of an image guided irradiation with a variable collimator with minimized exposure to non-targeted tissues and organs. Precise and accurate irradiation of lungs was carried out at either 14 or 16 Gy at an approximate dose rate of 2.6 Gy/min. Though a low throughput model, it is amenable to change the field size on the spot. No damage to other non-targeted organs was observed in histopathological evaluation. There was no significant change in peripheral blood counts of irradiated mice in comparison to naïve mice. Femoral bone marrow cells had no damage in irradiated mice. As expected, damage to the targeted tissue was observed in the histopathological evaluation and non-targeted tissue was found normal. Regeneration and increase of cellularity and megakaryocytes on GT3 treatment was compared to significant loss of cellularity in saline group. Peak alveolitis was observed on day 14 post-PBI and protection from alveolitis by GT3 was noted. In irradiated lung tissue, thirty proteins were found to be differentially expressed but modulated by GT3 to reverse the effects of irradiation. We propose that possible mode of action of GT3 could be Angiopoietin 2-Tie2 pathway leading to AKT/ERK pathways resulting in disruption in cell survival/angiogenesis.
Collapse
Affiliation(s)
- Vidya P Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sasha Stone
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Shukla Biswas
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Neel Sharma
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
39
|
Osteopontin Predicts Three-Month Outcome in Stroke Patients Treated by Reperfusion Therapies. J Clin Med 2020; 9:jcm9124028. [PMID: 33322093 PMCID: PMC7763291 DOI: 10.3390/jcm9124028] [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] [Received: 11/24/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022] Open
Abstract
Establishing a prognosis at hospital admission after stroke is a major challenge. Inflammatory processes, hemostasis, vascular injury, and tissue remodeling are all involved in the early response to stroke. This study analyzes whether 22 selected biomarkers, sampled at admission, predict clinical outcomes in 153 stroke patients treated by thrombolysis and mechanical endovascular treatment (MET). Biomarkers were related to hemostasis (u-plasminogen activator/urokinase (uPA/urokinase), serpin E1/PAI-1, serpin C1/antithrombin-III, kallikrein 6/neurosin, alpha 2-macroglobulin), inflammation[myloperoxidase (MPO), chemokine ligand 2/monocyte chemoattractant protein-1 chemokine (CCL2/MCP-1), adiponectin, resistin, cell-free DNA (cDNA), CD40 Ligand (CD40L)], endothelium activation (Vascular cell adhesion protein 1 (VCAM-1) intercellular adhesion molecule 1 (ICAM-1), platelet endothelial cell adhesion molecule 1 (CD31/PECAM-1)], and tissue remodeling (total cathepsin S, osteopontin, cystatin C, neuropilin-1, matrix metallopeptidase 2 (MMP-2), matrix metallopeptidase 3 (MMP-3), matrix metallopeptidase 9 (MMP-9), matrix metallopeptidase 13 (MMP-13)]. Correlations between their levels and excellent neurological improvement (ENI) at 24 h and good outcomes (mRS 0-2) at three months were tested. Osteopontin and favorable outcomes reached the significance level (p = 0.008); the adjusted OR per SD increase in log-transformed osteopontin was 0.34 (95%CI, 0.18-0.62). The relationship between total cathepsin S and MPO with ENI, was borderline of significance (p = 0.064); the adjusted OR per SD increase in log-transformed of total cathepsin S and MPO was 0.54 (95%CI, 0.35-0.81) and 0.51 (95%CI, 0.32-0.80), respectively. In conclusion, osteopontin levels predicted three-month favorable outcomes, supporting the use of this biomarker as a complement of clinical and radiological parameters for predicting stroke prognosis.
Collapse
|
40
|
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: 39] [Impact Index Per Article: 7.8] [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
|
41
|
Arnhold J. The Dual Role of Myeloperoxidase in Immune Response. Int J Mol Sci 2020; 21:E8057. [PMID: 33137905 PMCID: PMC7663354 DOI: 10.3390/ijms21218057] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
The heme protein myeloperoxidase (MPO) is a major constituent of neutrophils. As a key mediator of the innate immune system, neutrophils are rapidly recruited to inflammatory sites, where they recognize, phagocytose, and inactivate foreign microorganisms. In the newly formed phagosomes, MPO is involved in the creation and maintenance of an alkaline milieu, which is optimal in combatting microbes. Myeloperoxidase is also a key component in neutrophil extracellular traps. These helpful properties are contrasted by the release of MPO and other neutrophil constituents from necrotic cells or as a result of frustrated phagocytosis. Although MPO is inactivated by the plasma protein ceruloplasmin, it can interact with negatively charged components of serum and the extracellular matrix. In cardiovascular diseases and many other disease scenarios, active MPO and MPO-modified targets are present in atherosclerotic lesions and other disease-specific locations. This implies an involvement of neutrophils, MPO, and other neutrophil products in pathogenesis mechanisms. This review critically reflects on the beneficial and harmful functions of MPO against the background of immune response.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, 04 107 Leipzig, Germany
| |
Collapse
|
42
|
Huang C, Dong J, Jin X, Ma H, Zhang D, Wang F, Cheng L, Feng Y, Xiong X, Jiang J, Hu L, Lei M, Wu B, Zhang G. Intestinal anti-inflammatory effects of fuzi-ganjiang herb pair against DSS-induced ulcerative colitis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:112951. [PMID: 32574670 DOI: 10.1016/j.jep.2020.112951] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/21/2020] [Accepted: 05/03/2020] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzi and ganjiang are widely used as traditional Chinese medicines (TCM) in China, Korea, Japan, and many other southeast Asian countries for treating ulcerative colitis (UC), emesis and heart failure for more than 1800 years. However, the underlying mechanism of fuzi, ganjiang and fuzi-ganjiang herb pair is still unclear. In our study, we explored the therapeutic effects of fuzi, ganjiang and fuzi-ganjiang herb pair against dextran sulfate sodium (DSS)-induced UC in mice model, along with the relevant mechanism. MATERIALS AND METHODS The contents of each marker compound in fuzi decoction (FD), ganjiang decoction (GD) and fuzi-ganjiang decoction (FGD) were determined using LC-MS/MS. During the experiment, bodyweight changes in each group were monitored every 5 days. On the day of sacrifice, colonic length, disease activity index (DAI) and spleen weight were also evaluated and histopathological examination was performed through hematoxylin & eosin (H&E) staining. The levels of myeloperoxidase (MPO) and inflammatory cytokines in colon tissues were determined by enzyme-linked immunosorbent assay (ELISA), and then the relative mRNA productions of inflammatory mediators, such as MPO, inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 were measured by real-time polymerase chain reaction (PCR). Involvement of MAPK, STAT3 and NF-κB signaling pathways in the pathogenesis of UC was determined in each group using Western Blot (WB) analysis. RESULTS Compared with fuzi and ganjiang single decoction, the content of the alkaloids derived from fuzi (especially the diester alkaloid with strong toxicity, hypaconitine) in fuzi-ganjiang herb pair decoction was reduced. Additionally, the 6-gingerol, which was not found in ganjiang single decoction, was retained in fuzi-ganjiang herb pair decoction. FD, GD, and FGD significantly restored the bodyweight reduction, colon shortening, DAI elevation, splenomegaly and histological score in DSS-induced UC mice. Furthermore, except for the failure of low dosage of ganjiang decoction (GD-L) on IL-17A, all FD, GD and FGD significantly inhibited the production of MPO and inflammatory cytokines, such as IFN-γ, TNF-α, IL-1β, IL-6, IL-10 and IL-17A, and suppressed the relative expression of inflammatory mediators, such as MPO, iNOS and COX-2 mRNA in colon tissues of DSS-induced mice. According to WB analysis, fuzi, ganjiang and fuzi-ganjiang combination inhibited the activation of MAPK, NF-κB and STAT3 signaling pathways. CONCLUSIONS Our study demonstrated that fuzi, ganjiang and fuzi-ganjiang combination possess prominent anti-inflammatory activities against DSS-induced UC mice; the involved mechanism may be related to inhibition the activation of MAPK, NF-κB, and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Chuanqi Huang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Junli Dong
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Xiaoqi Jin
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China; College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Haoran Ma
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Dan Zhang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Fuqian Wang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Lu Cheng
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Yan Feng
- Department of Pathology, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Xin Xiong
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Jie Jiang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Lei Hu
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Mi Lei
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Bin Wu
- Department of Transfusion Medicine, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China
| | - Geng Zhang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional and Western Medicine), Wuhan, China.
| |
Collapse
|
43
|
Morris G, Puri BK, Olive L, Carvalho A, Berk M, Walder K, Gustad LT, Maes M. Endothelial dysfunction in neuroprogressive disorders-causes and suggested treatments. BMC Med 2020; 18:305. [PMID: 33070778 PMCID: PMC7570030 DOI: 10.1186/s12916-020-01749-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Potential routes whereby systemic inflammation, oxidative stress and mitochondrial dysfunction may drive the development of endothelial dysfunction and atherosclerosis, even in an environment of low cholesterol, are examined. MAIN TEXT Key molecular players involved in the regulation of endothelial cell function are described, including PECAM-1, VE-cadherin, VEGFRs, SFK, Rho GEF TRIO, RAC-1, ITAM, SHP-2, MAPK/ERK, STAT-3, NF-κB, PI3K/AKT, eNOS, nitric oxide, miRNAs, KLF-4 and KLF-2. The key roles of platelet activation, xanthene oxidase and myeloperoxidase in the genesis of endothelial cell dysfunction and activation are detailed. The following roles of circulating reactive oxygen species (ROS), reactive nitrogen species and pro-inflammatory cytokines in the development of endothelial cell dysfunction are then described: paracrine signalling by circulating hydrogen peroxide, inhibition of eNOS and increased levels of mitochondrial ROS, including compromised mitochondrial dynamics, loss of calcium ion homeostasis and inactivation of SIRT-1-mediated signalling pathways. Next, loss of cellular redox homeostasis is considered, including further aspects of the roles of hydrogen peroxide signalling, the pathological consequences of elevated NF-κB, compromised S-nitrosylation and the development of hypernitrosylation and increased transcription of atherogenic miRNAs. These molecular aspects are then applied to neuroprogressive disorders by considering the following potential generators of endothelial dysfunction and activation in major depressive disorder, bipolar disorder and schizophrenia: NF-κB; platelet activation; atherogenic miRs; myeloperoxidase; xanthene oxidase and uric acid; and inflammation, oxidative stress, nitrosative stress and mitochondrial dysfunction. CONCLUSIONS Finally, on the basis of the above molecular mechanisms, details are given of potential treatment options for mitigating endothelial cell dysfunction and activation in neuroprogressive disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | | | - Lisa Olive
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- School of Psychology, Faculty of Health, Deakin University, Geelong, Australia
| | - Andre Carvalho
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia.
- Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Ken Walder
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Lise Tuset Gustad
- Department of Circulation and medical imaging, Norwegian University of Technology and Science (NTNU), Trondheim, Norway
- Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Michael Maes
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
44
|
Lee DH, Yao C, Bhan A, Schlaeger T, Keefe J, Rodriguez BAT, Hwang SJ, Chen MH, Levy D, Johnson AD. Integrative Genomic Analysis Reveals Four Protein Biomarkers for Platelet Traits. Circ Res 2020; 127:1182-1194. [PMID: 32781905 PMCID: PMC8411925 DOI: 10.1161/circresaha.119.316447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
RATIONALE Mean platelet volume (MPV) and platelet count (PLT) are platelet measures that have been linked to cardiovascular disease (CVD) and mortality risk. Identifying protein biomarkers for these measures may yield insights into CVD mechanisms. OBJECTIVE We aimed to identify causal protein biomarkers for MPV and PLT among 71 CVD-related plasma proteins measured in FHS (Framingham Heart Study) participants. METHODS AND RESULTS We conducted integrative analyses of genetic variants associated with PLT/MPV with protein quantitative trait locus variants associated with plasma proteins followed by Mendelian randomization to infer causal relations of proteins for PLT/MPV. We also tested protein-PLT/MPV association in FHS participants. Using induced pluripotent stem cell-derived megakaryocyte clones that produce functional platelets, we conducted RNA-sequencing and analyzed expression differences between low- and high-platelet producing clones. We then performed small interfering RNA gene knockdown experiments targeting genes encoding proteins with putatively causal platelet effects in megakaryocyte clones to examine effects on platelet production. In protein-trait association analyses, ten proteins were associated with MPV and 31 with PLT. Mendelian randomization identified 4 putatively causal proteins for MPV and 4 for PLT. GP-5 (Glycoprotein V), GRN (granulin), and MCAM (melanoma cell adhesion molecule) were associated with PLT, while MPO (myeloperoxidase) showed significant association with MPV in both analyses. RNA-sequencing analysis results were directionally concordant with observed and Mendelian randomization-inferred associations for GP-5, GRN, and MCAM. In siRNA gene knockdown experiments, silencing GP-5, GRN, and MPO decreased PLTs. Genome-wide association study results suggest several of these may be linked to CVD risk. CONCLUSIONS We identified 4 proteins that are causally linked to PLTs. These proteins may also have roles in the pathogenesis of CVD via a platelet/blood coagulation-based mechanism.
Collapse
Affiliation(s)
- Dong Heon Lee
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Chen Yao
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | | | | | - Joshua Keefe
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Benjamin A T Rodriguez
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Ming-Huei Chen
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| | - Andrew D Johnson
- The Framingham Heart Study, Framingham, MA (D.H.L., C.Y., J.K., B.A.T.R., S,-J.H., M.-H.C., D.L., A.D.J.)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute National Institutes of Health, Bethesda, MD (D.H.L., C.Y., J.K., B.A.T.R., S.-J.H., M.-H.C., D.L., A.D.J.)
| |
Collapse
|
45
|
Klinke A, Schubert T, Müller M, Legchenko E, Zelt JGE, Shimauchi T, Napp LC, Rothman AMK, Bonnet S, Stewart DJ, Hansmann G, Rudolph V. Emerging therapies for right ventricular dysfunction and failure. Cardiovasc Diagn Ther 2020; 10:1735-1767. [PMID: 33224787 PMCID: PMC7666928 DOI: 10.21037/cdt-20-592] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Therapeutic options for right ventricular (RV) dysfunction and failure are strongly limited. Right heart failure (RHF) has been mostly addressed in the context of pulmonary arterial hypertension (PAH), where it is not possible to discern pulmonary vascular- and RV-directed effects of therapeutic approaches. In part, opposing pathomechanisms in RV and pulmonary vasculature, i.e., regarding apoptosis, angiogenesis and proliferation, complicate addressing RHF in PAH. Therapy effective for left heart failure is not applicable to RHF, e.g., inhibition of adrenoceptor signaling and of the renin-angiotensin system had no or only limited success. A number of experimental studies employing animal models for PAH or RV dysfunction or failure have identified beneficial effects of novel pharmacological agents, with most promising results obtained with modulators of metabolism and reactive oxygen species or inflammation, respectively. In addition, established PAH agents, in particular phosphodiesterase-5 inhibitors and soluble guanylate cyclase stimulators, may directly address RV integrity. Promising results are furthermore derived with microRNA (miRNA) and long non-coding RNA (lncRNA) blocking or mimetic strategies, which can target microvascular rarefaction, inflammation, metabolism or fibrotic and hypertrophic remodeling in the dysfunctional RV. Likewise, pre-clinical data demonstrate that cell-based therapies using stem or progenitor cells have beneficial effects on the RV, mainly by improving the microvascular system, however clinical success will largely depend on delivery routes. A particular option for PAH is targeted denervation of the pulmonary vasculature, given the sympathetic overdrive in PAH patients. Finally, acute and durable mechanical circulatory support are available for the right heart, which however has been tested mostly in RHF with concomitant left heart disease. Here, we aim to review current pharmacological, RNA- and cell-based therapeutic options and their potential to directly target the RV and to review available data for pulmonary artery denervation and mechanical circulatory support.
Collapse
Affiliation(s)
- Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Torben Schubert
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Marion Müller
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Jason G. E. Zelt
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - L. Christian Napp
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - Duncan J. Stewart
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Volker Rudolph
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
46
|
El Kazzi M, Rayner BS, Chami B, Dennis JM, Thomas SR, Witting PK. Neutrophil-Mediated Cardiac Damage After Acute Myocardial Infarction: Significance of Defining a New Target Cell Type for Developing Cardioprotective Drugs. Antioxid Redox Signal 2020; 33:689-712. [PMID: 32517486 PMCID: PMC7475094 DOI: 10.1089/ars.2019.7928] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Significance: Acute myocardial infarction (AMI) is a leading cause of death worldwide. Post-AMI survival rates have increased with the introduction of angioplasty as a primary coronary intervention. However, reperfusion after angioplasty represents a clinical paradox, restoring blood flow to the ischemic myocardium while simultaneously inducing ion and metabolic imbalances that stimulate immune cell recruitment and activation, mitochondrial dysfunction and damaging oxidant production. Recent Advances: Preclinical data indicate that these metabolic imbalances contribute to subsequent heart failure through sustaining local recruitment of inflammatory leukocytes and oxidative stress, cardiomyocyte death, and coronary microvascular disturbances, which enhance adverse cardiac remodeling. Both left ventricular dysfunction and heart failure are strongly linked to inflammation and immune cell recruitment to the damaged myocardium. Critical Issues: Overall, therapeutic anti-inflammatory and antioxidant agents identified in preclinical trials have failed in clinical trials. Future Directions: The versatile neutrophil-derived heme enzyme, myeloperoxidase (MPO), is gaining attention as an important oxidative mediator of reperfusion injury, vascular dysfunction, adverse ventricular remodeling, and atrial fibrillation. Accordingly, there is interest in therapeutically targeting neutrophils and MPO activity in the setting of heart failure. Herein, we discuss the role of post-AMI inflammation linked to myocardial damage and heart failure, describe previous trials targeting inflammation and oxidative stress post-AMI, highlight the potential adverse impact of neutrophil and MPO, and detail therapeutic options available to target MPO clinically in AMI patients.
Collapse
Affiliation(s)
- Mary El Kazzi
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | | | - Belal Chami
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Joanne Marie Dennis
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Shane Ross Thomas
- Department of Pathology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Paul Kenneth Witting
- Discipline of Pathology, Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
47
|
Vanichkitrungruang S, Chuang CY, Hawkins CL, Davies MJ. Myeloperoxidase-derived damage to human plasma fibronectin: Modulation by protein binding and thiocyanate ions (SCN -). Redox Biol 2020; 36:101641. [PMID: 32863239 PMCID: PMC7378696 DOI: 10.1016/j.redox.2020.101641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/29/2022] Open
Abstract
Endothelial cell dysfunction is an early event in cardiovascular disease and atherosclerosis. The origin of this dysfunction is unresolved, but accumulating evidence implicates damaging oxidants, including hypochlorous acid (HOCl), a major oxidant produced by myeloperoxidase (MPO), during chronic inflammation. MPO is released extracellularly by activated leukocytes and binds to extracellular molecules including fibronectin, a major matrix glycoprotein involved in endothelial cell binding. We hypothesized that MPO binding might influence the modifications induced on fibronectin, when compared to reagent HOCl, with this including alterations to the extent of damage to protein side-chains, modified structural integrity, changes to functional domains, and impact on naïve human coronary artery endothelial cell (HCAEC) adhesion and metabolic activity. The effect of increasing concentrations of the alternative MPO substrate thiocyanate (SCN-), which might decrease HOCl formation were also examined. Exposure of fibronectin to MPO/H2O2/Cl- is shown to result in damage to the functionally important cell-binding and heparin-binding fragments, gross structural changes to the protein, and altered HCAEC adhesion and activity. Differences were observed between stoichiometric, and above-stoichiometric MPO concentrations consistent with an effect of MPO binding to fibronectin. In contrast, MPO/H2O2/SCN- induced much less marked changes and limited protein damage. Addition of increasing SCN- concentrations to the MPO/H2O2/Cl- system provided protection, with 20 μM of this anion rescuing damage to functionally-important domains, decreasing chemical modification, and maintaining normal HCAEC behavior. Modulating MPO binding to fibronectin, or enhancing SCN- levels at sites of inflammation may therefore limit MPO-mediated damage, and be of therapeutic value.
Collapse
Affiliation(s)
- Siriluck Vanichkitrungruang
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Michael J Davies
- The Heart Research Institute, Newtown, NSW, Australia; Faculty of Medicine, The University of Sydney, NSW, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
48
|
Li Y, Lin P, Wang S, Li S, Wang R, Yang L, Wang H. Quantitative analysis of differentially expressed proteins in psoriasis vulgaris using tandem mass tags and parallel reaction monitoring. Clin Proteomics 2020; 17:30. [PMID: 32817748 PMCID: PMC7425065 DOI: 10.1186/s12014-020-09293-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/06/2020] [Indexed: 12/29/2022] Open
Abstract
Background Psoriasis vulgaris (PV) is a chronic autoimmune inflammatory disease with epidermal hyperkeratosis and parakeratosis. Methods The study was to elucidate the pathogenesis of PV by quantitative proteomic analysis of skin lesion biopsies of PV and healthy tissues with tandem mass tags (TMTs) coupled with liquid chromatography–mass spectrometry (LC–MS)/MS. Results A total of 4562 differentially expressed proteins (DEPs) between PV lesional tissues (n = 11) and healthy tissues (n = 11) were identified, of which 299 were upregulated and 206 were downregulated using |fold change| > 1.3 as the cutoff threshold. The Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that the DEPs were mainly enriched in the activation of immune cells (drug metabolism pathway, NOD-like pathway, and IL-17 pathway), cell proliferation (ribosomal pathway, DNA replication pathway, and base replication pathway), metabolism-related pathways (fatty acid biosynthesis and metabolism, PPAR pathway, glycerophospholipid metabolism, and cortisol synthesis and breakdown), and glandular secretion (saliva secretion, gastric acid secretion, and pancreatic fluid secretion). Thirteen DEPs that were relatively highly expressed in the drug metabolism pathway were validated with parallel reaction monitoring (PRM), of which MPO, TYMP, IMPDH2, GSTM4, and ALDH3A1 were highly expressed in PV, whereas CES1, MAOB, MGST1, and GSTT1 were less expressed in PV. Conclusions These findings confirmed that these proteins participate in the drug metabolism-other enzyme pathways and play crucial roles in the activation and proliferation of immune cells in the pathogenesis of PV.
Collapse
Affiliation(s)
- Yu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Peng Lin
- Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong People's Republic of China
| | - Siyao Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shuang Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Rui Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Lin Yang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| | - Hongmei Wang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| |
Collapse
|
49
|
Mishra OP, Popov AV, Pietrofesa RA, Hwang WT, Andrake M, Nakamaru-Ogiso E, Christofidou-Solomidou M. Radiation activates myeloperoxidase (MPO) to generate active chlorine species (ACS) via a dephosphorylation mechanism - inhibitory effect of LGM2605. Biochim Biophys Acta Gen Subj 2020; 1864:129548. [PMID: 32035161 PMCID: PMC8413008 DOI: 10.1016/j.bbagen.2020.129548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/16/2020] [Accepted: 02/03/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Radiation exposure of tissues is associated with inflammatory cell influx. Myeloperoxidase (MPO) is an enzyme expressed in granulocytes, such as neutrophils (PMN) and macrophages, responsible for active chlorine species (ACS) generation. The present study aimed to: 1) determine whether exposure to γ-irradiation induces MPO-dependent ACS generation in murine PMN; 2) elucidate the mechanism of radiation-induced ACS generation; and 3) evaluate the effect of the synthetic lignan LGM2605, known for ACS scavenging properties. METHODS MPO-dependent ACS generation was determined by using hypochlorite-specific 3'-(p-aminophenyl) fluorescein (APF) and a highly potent MPO inhibitor, 4-aminobenzoic acid hydrazide (ABAH), and confirmed in PMN derived from MPO-/- mice. Radiation-induced MPO activation was determined by EPR spectroscopy and computational analysis identified tyrosine, serine, and threonine residues near MPO's active site. RESULTS γ-radiation increased MPO-dependent ACS generation dose-dependently in human MPO and in wild-type murine PMN, but not in PMN from MPO-/- mice. LGM2605 decreased radiation-induced, MPO-dependent ACS. Protein tyrosine phosphatase (PTP) and protein serine/threonine phosphatase (PSTP) inhibitors decreased the radiation-induced increase in ACS. Peroxidase cycle results demonstrate that tyrosine phosphorylation blocks MPO Compound I formation by preventing catalysis on H2O2 in the active site of MPO. EPR data demonstrate that γ-radiation increased tyrosyl radical species formation in a dose-dependent manner. CONCLUSIONS We demonstrate that γ-radiation induces MPO-dependent generation of ACS, which is dependent, at least in part, by protein tyrosine and Ser/Thr dephosphorylation and is reduced by LGM2605. This study identified for the first time a novel protein dephosphorylation-dependent mechanism of radiation-induced MPO activation.
Collapse
Affiliation(s)
- Om P Mishra
- University of Pennsylvania Perelman School of Medicine, Department of Medicine, Pulmonary, Allergy, and Critical Care Division, Philadelphia, PA 19104, United States of America.
| | - Anatoliy V Popov
- University of Pennsylvania Perelman School of Medicine, Department of Radiology, Philadelphia, PA 19104, United States of America.
| | - Ralph A Pietrofesa
- University of Pennsylvania Perelman School of Medicine, Department of Medicine, Pulmonary, Allergy, and Critical Care Division, Philadelphia, PA 19104, United States of America.
| | - Wei-Ting Hwang
- University of Pennsylvania Perelman School of Medicine, Department of Biostatistics, Epidemiology, and Informatics, Philadelphia, PA 19104, United States of America.
| | - Mark Andrake
- Molecular Modeling Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, United States of America.
| | - Eiko Nakamaru-Ogiso
- Children's Hospital of Philadelphia, Department of Pediatrics, Philadelphia, PA 19104, United States of America.
| | - Melpo Christofidou-Solomidou
- University of Pennsylvania Perelman School of Medicine, Department of Medicine, Pulmonary, Allergy, and Critical Care Division, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
50
|
Wang LF, Li Y, Landsittel DP, Reis SE, Levesque MC, Jones DM, Gartland R, Avolio J, Shoushtari A, Qi Z, Dezfulian C, Moreland LW, Liang KP. Identifying Vulnerable Plaque in Rheumatoid Arthritis Using Novel Microbubble Contrast-Enhanced Carotid Ultrasonography and Serum Biomarkers. JOURNAL OF DIAGNOSTIC MEDICAL SONOGRAPHY 2020. [DOI: 10.1177/8756479320922512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective: Rheumatoid arthritis (RA) is associated with increased risk of cardiovascular disease. Adventitial vasa vasorum density (aVVD), the vessel density of the vasa vasorum, is a surrogate measure for atherosclerotic plaque vulnerability. The purpose of this study was to compare the adventitial vasa vasorum density (aVVD) in RA and non-RA control participants using novel carotid artery contrast-enhanced ultrasound (CEUS). In addition, we investigate associations of aVVD with traditional cardiovascular (CV) risk factors, vascular and inflammatory biomarkers, and RA disease activity. Methods: The study was a cross-sectional analysis of patients with RA and control participants without RA or other autoimmune disease. CV disease risk, biomarkers, and CEUS images were collected on all patients. Results: aVVD was quantified in 86 patients with RA and 95 non-RA control participants. Nitrite, CD40L, E-selectin, matrix metalloproteinase 9, intercellular adhesion molecule 1, vascular cell adhesion molecule 1, myeloperoxidase (MPO), high-sensitivity C-reactive protein (hsCRP), and erythrocyte sedimentation rate were measured. Median aVVD was higher in patients with RA (0.59 [0.47–0.69] vs 0.64 [0.54–0.62]; P = .02). In patients with RA, MPO was lower (253.5 [153.2–480] vs 470.8 [274.2–830.1] ng/mL; P = .0002) and ESR was higher (15.5 [11–25] vs 13 [9–20] mm/h; P = .02). aVVD was correlated with MPO ( r = −0.33, P = .001) and hsCRP ( r = 0.25, P = .02) in control participants only, associations that remained significant after adjusting for number of CV risk factors and age. No significant correlations were found between aVVD and RA disease activity measures. Conclusions: Using a novel application of CEUS, we found that aVVD, an early measure of plaque vulnerability, was significantly higher in RA than control subjects, even after adjusting for CV risk factors. Differences in correlation of aVVD with vascular biomarkers and CV risk factors suggest RA-related differences in atherosclerotic progression.
Collapse
Affiliation(s)
- Linda F. Wang
- School of Medicine, University of Pittsburgh, Pittsburgh PA, USA
| | - Yaming Li
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh PA, USA
| | | | - Steven E. Reis
- Division of Cardiology, University of Pittsburgh, Pittsburgh PA, USA
| | - Marc C. Levesque
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Donald M. Jones
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Rachel Gartland
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Jennifer Avolio
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh PA, USA
| | - Ali Shoushtari
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh PA, USA
| | - Zengbiao Qi
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Cameron Dezfulian
- Vascular Medicine Institute and Critical Care Medicine, University of Pittsburgh, Pittsburgh PA, USA
| | - Larry W. Moreland
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Kimberly P. Liang
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|