1
|
Ricart K, McCommis KS, Ford DA, Patel RP. 2-Chloro- and 2-Bromopalmitic acids inhibit mitochondrial function in airway epithelial cells. ADVANCES IN REDOX RESEARCH 2025; 14:100118. [PMID: 40123941 PMCID: PMC11928162 DOI: 10.1016/j.arres.2024.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
2-Chloropalmitic acid (2-ClPA) and 2-bromopalmitic acid (2-BrPa) increase in inflammatory lung disease associated with formation of hypochlorous or hypobromous acid, and exposure to halogen gases. Moreover, these lipids may elicit cell responses that contribute to lung injury, but the mechanisms remain unclear. Here, we tested the hypothesis that 2-ClPA and 2-BrPA induce metabolic defects in airway epithelial cells by targeting mitochondria. H441 or primary human airway epithelial cells were treated with 2-ClPA or 2-BrPA and bioenergetics measured using oxygen consumption rates and extracellular acidification rates, as well as respiratory complex activities. Relative to vehicle or palmitic acid, both 2-halofatty acids inhibited ATP-linked oxygen consumption and reserve capacity, suggestive of increased proton leak. However, neither 2-ClPA nor 2-BrPA altered mitochondrial membrane potential, suggesting proton leak does not underlie inhibited ATP-linked oxygen consumption. Interestingly, complex II activity was significantly inhibited which may contribute to diminished reserve capacity, but activity of complexes I, III and IV remain unchanged. Taken together, the presented data highlight the potential of 2-halofatty acids to disrupt bioenergetics and in turn cause cellular dysfunction.
Collapse
Affiliation(s)
- Karina Ricart
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kyle S. McCommis
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - David A. Ford
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Rakesh P. Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
2
|
Yang K, Lu Y, Gu J, Nie Y, Zhang T. Identifying novel aging-related diagnostic and prognostic models and aging-targeted drugs for sepsis patients. Sci Rep 2024; 14:31445. [PMID: 39732977 PMCID: PMC11682178 DOI: 10.1038/s41598-024-83111-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Sepsis is defined as a dysfunctional, life-threatening response to infection leading to multiorgan dysfunction and failure. During the past decade, studies have highlighted the relationship between sepsis and aging. However, the role of aging-related mechanisms in the progression and prognosis of sepsis remains unclear. In the present study, we divided sepsis patients into High- and Low-aging groups based on the gene set variation analysis (GSVA) scores of GOBP-AGING gene set. Sepsis patients in the high-aging group exhibited higher levels of infiltration of innate immune cells, lower levels of infiltration of adaptive immune cells, and a worse prognosis than those in the Low-aging group. Additionally, the MPO to MME ratio (MPO/MME) appears to be an effective biomarker for predicting the prognosis of sepsis patients. Moreover, ARG1/SEC63 and ARG1/CDKN1C appear to be effective and robust biomarkers for the early diagnosis of sepsis patients. Finally, we found that thalidomide (TAL) significantly ameliorated LPS induced inflammation and organ injury and attenuated LPS induced cellular senescence in lung and kidney. Overall, this study provides new insights into the heterogeneity of sepsis, reveals the vital role of aging-related markers in the prognosis and diagnosis of sepsis and demonstrates that TAL is a novel aging-targeted drug for sepsis patients by attenuating LPS induced cellular senescence.
Collapse
Affiliation(s)
- Kai Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yaoyao Lu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jian Gu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yingli Nie
- Department of Dermatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China.
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Lee T, Lee J, Shin DH, Lee H, Kim SK. Prognostic and Diagnostic Power of Delta Neutrophil Index and Mean Platelet Component in Febrile Patients with Suspected Sepsis. Biomedicines 2023; 11:3190. [PMID: 38137411 PMCID: PMC10740452 DOI: 10.3390/biomedicines11123190] [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: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The delta neutrophil index (DNI), a prognostic and diagnostic marker for sepsis, is based on the leukocyte count. Platelet activation, similar to leukocyte activation, plays a crucial role in host defense against pathogens and may serve as a predictor of sepsis outcome. However, the combined evaluation of mean platelet component (MPC) and DNI has rarely been used to assess sepsis. METHODS To assess the prognostic and diagnostic validity of the simultaneous evaluation of DNI and MPC in cases of human febrile sepsis, we conducted measurements of cellular indices, including DNI and MPC, as well as molecular biomarkers, including procalcitonin (PCT) and C-reactive protein (CRP). This study was carried out in patients admitted to the emergency department with suspected sepsis. RESULTS Using a cutoff value of 2.65%, the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of the DNI in sepsis were found to be 69%, 73.9%, 77.9%, and 64.1%, respectively. Furthermore, significant differences in DNI and MPC levels were observed between the sepsis and non-sepsis groups (6.7 ± 7.8% versus 2.1 ± 2.2% (p = 0.000) and 26.0 ± 1.9 g/dL versus 26.8 ± 1.4 g/dL (p = 0.002), respectively). Notably, there was a negative correlation between DNI and MPC, with the strength of the correlation varying based on the cause of sepsis. By setting the cutoff value of the DNI to 6.2%, its sensitivity, specificity, and NPV improved to 100%, 80.3%, and 100%, respectively, although the PPV remained at 10.6%. CONCLUSIONS In our study, the DNI demonstrates superior effectiveness compared with other molecular biomarkers, such as CRP and procalcitonin, in distinguishing septic febrile patients from non-septic febrile patients. Additionally, a negative correlation exists between MPC and DNI, making MPC a valuable marker for differentiating the etiology of sepsis. These findings hold significant clinical implications, as DNI/MPC evaluation is a cost-effective and readily applicable approach in various impending sepsis scenarios. Notably, this study represents the first examination of the prognostic and diagnostic validity of employing the simultaneous evaluation of DNI and MPC in human cases of febrile sepsis.
Collapse
Affiliation(s)
- Taehun Lee
- Department of Emergency Medicine, College of Medicine, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Republic of Korea;
| | - Jongwook Lee
- Department of Laboratory Medicine, Konyang University Hospital, Daejeon 35465, Republic of Korea;
| | - Dong Hoon Shin
- Department of Laboratory Medicine, College of Medicine, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Republic of Korea;
| | - Hyungdon Lee
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Republic of Korea
| | - Soo-Ki Kim
- Department of Microbiology, Wonju College of Medicine, Research Institute of Metabolism and Inflammation Research, Yonsei University, Wonju 26426, Republic of Korea
| |
Collapse
|
4
|
Shen S, Wang X, Zhu X, Rasam S, Ma M, Huo S, Qian S, Zhang M, Qu M, Hu C, Jin L, Tian Y, Sethi S, Poulsen D, Wang J, Tu C, Qu J. High-quality and robust protein quantification in large clinical/pharmaceutical cohorts with IonStar proteomics investigation. Nat Protoc 2023; 18:700-731. [PMID: 36494494 PMCID: PMC10673696 DOI: 10.1038/s41596-022-00780-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 09/22/2022] [Indexed: 12/14/2022]
Abstract
Robust, reliable quantification of large sample cohorts is often essential for meaningful clinical or pharmaceutical proteomics investigations, but it is technically challenging. When analyzing very large numbers of samples, isotope labeling approaches may suffer from substantial batch effects, and even with label-free methods, it becomes evident that low-abundance proteins are not reliably measured owing to unsufficient reproducibility for quantification. The MS1-based quantitative proteomics pipeline IonStar was designed to address these challenges. IonStar is a label-free approach that takes advantage of the high sensitivity/selectivity attainable by ultrahigh-resolution (UHR)-MS1 acquisition (e.g., 120-240k full width at half maximum at m/z = 200) which is now widely available on ultrahigh-field Orbitrap instruments. By selectively and accurately procuring quantitative features of peptides within precisely defined, very narrow m/z windows corresponding to the UHR-MS1 resolution, the method minimizes co-eluted interferences and substantially enhances signal-to-noise ratio of low-abundance species by decreasing noise level. This feature results in high sensitivity, selectivity, accuracy and precision for quantification of low-abundance proteins, as well as fewer missing data and fewer false positives. This protocol also emphasizes the importance of well-controlled, robust experimental procedures to achieve high-quality quantification across a large cohort. It includes a surfactant cocktail-aided sample preparation procedure that achieves high/reproducible protein/peptide recoveries among many samples, and a trapping nano-liquid chromatography-mass spectrometry strategy for sensitive and reproducible acquisition of UHR-MS1 peptide signal robustly across a large cohort. Data processing and quality evaluation are illustrated using an example dataset ( http://proteomecentral.proteomexchange.org ), and example results from pharmaceutical project and one clinical project (patients with acute respiratory distress syndrome) are shown. The complete IonStar pipeline takes ~1-2 weeks for a sample cohort containing ~50-100 samples.
Collapse
Affiliation(s)
- Shichen Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Xue Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Xiaoyu Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sailee Rasam
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Min Ma
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shihan Huo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Shuo Qian
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ming Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Miao Qu
- Department of Neurology, Xuanwu Hospital, Beijing, China
| | - Chenqi Hu
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Liang Jin
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Yu Tian
- AbbVie Bioresearch Center, Worcester, MA, USA
| | - Sanjay Sethi
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - David Poulsen
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Chengjian Tu
- BioProduction Group, Thermo Fisher Scientific, Buffalo, NY, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
5
|
Shakya S, McGuffee RM, Ford DA. Characterization of N-Acetyl Cysteine Adducts with Exogenous and Neutrophil-Derived 2-Chlorofatty Aldehyde. Antioxidants (Basel) 2023; 12:antiox12020504. [PMID: 36830062 PMCID: PMC9952649 DOI: 10.3390/antiox12020504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Hypochlorous acid is produced by leukocyte myeloperoxidase activity. 2-Chlorofatty aldehydes (2-ClFALDs) are formed when hypochlorous acid attacks the plasma membrane phospholipid plasmalogen molecular subclass and are thus produced following leukocyte activation as well as in the lungs of mice exposed to chlorine gas. The biological role of 2-ClFALD is largely unknown. Recently, we used an alkyne analog (2-ClHDyA) of the 2-ClFALD molecular species, 2-chlorohexadecanal (2-ClHDA), to identify proteins covalently modified by 2-ClHDyA in endothelial cells and epithelial cells. Here, we demonstrate that 2-ClHDA reduces the metabolic activity of RAW 264.7 cells in a dose-dependent manner. 2-ClHDyA localizes to the mitochondria, endoplasmic reticulum and Golgi in RAW 264.7 cells and modifies many proteins. The thiol-containing precursor of glutathione, N-acetyl cysteine (NAC), was shown to produce an adduct with 2-ClHDA with the loss of Cl- (HDA-NAC). This adduct was characterized in both positive and negative ion modes using LC-MS/MS and electrospray ionization. NAC treatment of neutrophils reduced the 2-ClFALD levels in PMA-stimulated cells with subsequent increases in HDA-NAC. NAC treatments reduced the 2-ClHDA-elicited loss of metabolic activity in RAW 264.7 cells as well as 2-ClHDA protein modification. These studies demonstrate that 2-ClFALD toxic effects can be reduced by NAC, which reduces protein modification.
Collapse
Affiliation(s)
- Shubha Shakya
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Reagan M. McGuffee
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - David A. Ford
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Correspondence:
| |
Collapse
|
6
|
Liu L, Chen X, Jiang Y, Yuan Y, Yang L, Hu Q, Tang J, Meng X, Xie C, Shen X. Brevilin A Ameliorates Acute Lung Injury and Inflammation Through Inhibition of NF-κB Signaling via Targeting IKKα/β. Front Pharmacol 2022; 13:911157. [PMID: 35774606 PMCID: PMC9237443 DOI: 10.3389/fphar.2022.911157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Acute lung injury (ALI) is life-threatening disease characterized by uncontrolled inflammatory response. IKKα/β, the key kinases in the activation of NF-κB pathway, are implicated in inflammatory pulmonary injury, and represent attractive targets for ALI therapy. Brevilin A (BVA) is a sesquiterpene lactone from Centipeda minima, a Chinese herb used to treat inflammatory diseases. This study aims to investigate the inhibition of BVA on ALI, with focus on clarifying the molecular mechanisms involved in BVA-mediated anti-inflammatory activity in macrophages. Briefly, BVA significantly inhibited the production of NO and PGE2 by suppressing iNOS and COX2 expression, and suppressed the mRNA expression of IL-1β, IL-6, and TNFα in LPS/IFNγ-stimulated RAW264.7 macrophages. The anti-inflammatory activity of BVA was further confirmed in LPS/IFNγ-stimulated BMDMs and TNFα/IFNγ-exposed RAW264.7 cells. In vivo, BVA effectively attenuated LPS-induced lung damage, inflammatory infiltration, and production of pro-inflammatory cytokines, including MPO, IL-1β, IL-6, TNFα, and PGE2. Mechanistically, BVA could covalently bind to the cysteine 114 of IKKα/β, and effectively inhibiting the activity and function of IKKα/β, thereby resulting in the suppression of phosphorylation and degradation of IκBα and the subsequent activation of NF-κB signaling. Furthermore, pretreatment of DTT, a thiol ligand donor, significantly abolished BVA-mediated effects in LPS/IFNγ-stimulated RAW264.7 cells, suggesting the crucial role of the electrophilic α, β-unsaturated ketone of BVA on its anti-inflammatory activity. These results suggest that BVA ameliorates ALI through inhibition of NF-κB signaling via covalently targeting IKKα/β, raising the possibility that BVA could be effective in the treatment of ALI and other diseases harboring aberrant NF-κB signaling.
Collapse
Affiliation(s)
- Lu Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Chen
- Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yifang Jiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Yuan
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luyao Yang
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunguang Xie
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunguang Xie, ; Xiaofei Shen,
| | - Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunguang Xie, ; Xiaofei Shen,
| |
Collapse
|
7
|
Endothelial Cell Protein Targeting by Myeloperoxidase-Derived 2-Chlorofatty Aldehyde. Antioxidants (Basel) 2022; 11:antiox11050940. [PMID: 35624804 PMCID: PMC9138145 DOI: 10.3390/antiox11050940] [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: 04/07/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
Neutrophils are important cellular mediators of injury and repair in diseases including ischemic heart disease, atherosclerosis, and sepsis. Myeloperoxidase-derived (MPO)-oxidants released from neutrophils are potential mediators of endothelial injury in disease. MPO-derived HOCl attacks plasmalogen phospholipid to liberate 2-chlorofatty aldehyde (2-ClFALD). Both 2-ClFALD and its oxidation product, 2-chlorofatty acid (2-ClFA), are electrophilic lipids, and both probably react with proteins through several mechanisms. In the present study, we investigate protein modification specifically by 2-ClFALD under non-reducing conditions (e.g., without stabilizing Schiff base bonds), which likely reflects nucleophilic targeting of the electrophilic chlorinated carbon. Protein modification by the ω-alkyne analog of 2-chlorohexadecanal (2-ClHDA), 2-ClHDyA, was compared to that with the ω-alkyne analog of 2-chlorohexadecanoic acid (2-ClHA), 2-ClHyA, in multiple cell lines, which demonstrated 2-ClFALD preferentially modifies proteins compared to 2-ClFA. The 2-ClHDyA modified proteins from EA.hy926 cells and human lung microvascular endothelial cells analyzed by shotgun proteomics and over-representation analysis included adherens junction, cell adhesion molecule binding, and cell substrate junction enrichment categories. It is possible that proteins in these groups may have roles in previously described 2-ClFALD-elicited endothelial barrier dysfunction.
Collapse
|
8
|
Pike DP, McGuffee RM, Geerling E, Albert CJ, Hoft DF, Shashaty MGS, Meyer NJ, Pinto AK, Ford DA. Plasmalogen Loss in Sepsis and SARS-CoV-2 Infection. Front Cell Dev Biol 2022; 10:912880. [PMID: 35784479 PMCID: PMC9242022 DOI: 10.3389/fcell.2022.912880] [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: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmalogens are plasma-borne antioxidant phospholipid species that provide protection as cellular lipid components during cellular oxidative stress. In this study we investigated plasma plasmalogen levels in human sepsis as well as in rodent models of infection. In humans, levels of multiple plasmenylethanolamine molecular species were decreased in septic patient plasma compared to control subject plasma as well as an age-aligned control subject cohort. Additionally, lysoplasmenylcholine levels were significantly decreased in septic patients compared to the control cohorts. In contrast, plasma diacyl phosphatidylethanolamine and phosphatidylcholine levels were elevated in septic patients. Lipid changes were also determined in rats subjected to cecal slurry sepsis. Plasma plasmenylcholine, plasmenylethanolamine, and lysoplasmenylcholine levels were decreased while diacyl phosphatidylethanolamine levels were increased in septic rats compared to control treated rats. Kidney levels of lysoplasmenylcholine as well as plasmenylethanolamine molecular species were decreased in septic rats. Interestingly, liver plasmenylcholine and plasmenylethanolamine levels were increased in septic rats. Since COVID-19 is associated with sepsis-like acute respiratory distress syndrome and oxidative stress, plasmalogen levels were also determined in a mouse model of COVID-19 (intranasal inoculation of K18 mice with SARS-CoV-2). 3 days following infection, lung infection was confirmed as well as cytokine expression in the lung. Multiple molecular species of lung plasmenylcholine and plasmenylethanolamine were decreased in infected mice. In contrast, the predominant lung phospholipid, dipalmitoyl phosphatidylcholine, was not decreased following SARS-CoV-2 infection. Additionally total plasmenylcholine levels were decreased in the plasma of SARS-CoV-2 infected mice. Collectively, these data demonstrate the loss of plasmalogens during both sepsis and SARS-CoV-2 infection. This study also indicates plasma plasmalogens should be considered in future studies as biomarkers of infection and as prognostic indicators for sepsis and COVID-19 outcomes.
Collapse
Affiliation(s)
- Daniel P Pike
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Reagan M McGuffee
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Carolyn J Albert
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Daniel F Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Michael G S Shashaty
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.,Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.,Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
9
|
Amunugama K, Jellinek MJ, Kilroy MP, Albert CJ, Rasi V, Hoft DF, Shashaty MGS, Meyer NJ, Ford DA. Identification of novel neutrophil very long chain plasmalogen molecular species and their myeloperoxidase mediated oxidation products in human sepsis. Redox Biol 2021; 48:102208. [PMID: 34902676 PMCID: PMC8671113 DOI: 10.1016/j.redox.2021.102208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/07/2021] [Indexed: 11/25/2022] Open
Abstract
Plasmalogens are a class of phospholipids containing vinyl ether linked aliphatic groups at the sn-1 position. Plasmalogens are known to contain 16- and 18-carbon aliphatic groups at the sn-1 position. Here, we reveal that the human neutrophil plasmenylethanolamine pool uniquely includes molecular species with very long carbon chain (VLC) aliphatic groups, including 20-, 22- and 24-carbon vinyl ether linked aliphatic groups at the sn-1 position. We identified these novel VLC plasmalogen species by electrospray ionization mass spectrometry methods. VLC plasmalogens were only found in the neutrophil plasmenylethanolamine pool. During neutrophil activation, VLC plasmenylethanolamines undergo myeloperoxidase-dependent oxidation to produce VLC 2-chlorofatty aldehyde and its oxidation product, 2-chlorofatty acid (2-ClFA). Furthermore, plasma concentrations of VLC 2-ClFA are elevated in human sepsis. These studies demonstrate for the first time VLC plasmenylethanolamine molecular species, their myeloperoxidase-mediated chlorolipid products and the presence of these chlorolipids in human sepsis.
Collapse
Affiliation(s)
- Kaushalya Amunugama
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Matthew J Jellinek
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Megan P Kilroy
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Carolyn J Albert
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Valerio Rasi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Daniel F Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Michael G S Shashaty
- Pulmonary, Allergy, Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA; Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104-6021, USA
| | - Nuala J Meyer
- Pulmonary, Allergy, Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA; Center for Translational Lung Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104-6021, USA
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA.
| |
Collapse
|
10
|
Gotts JE, Maishan M, Chun L, Fang X, Han C, Chiueh V, Khakoo AY, Lee T, Stolina M, Matthay MA. Delayed angiopoietin-2 blockade reduces influenza-induced lung injury and improves survival in mice. Physiol Rep 2021; 9:e15081. [PMID: 34755490 PMCID: PMC8578883 DOI: 10.14814/phy2.15081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/24/2022] Open
Abstract
Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.
Collapse
Affiliation(s)
- Jeffrey E. Gotts
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Mazharul Maishan
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lauren Chun
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Xiaohui Fang
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Chun‐Ya Han
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Venice Chiueh
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Aarif Y. Khakoo
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - TaeWeon Lee
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Marina Stolina
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Michael A. Matthay
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
11
|
Addis DR, Aggarwal S, Lazrak A, Jilling T, Matalon S. Halogen-Induced Chemical Injury to the Mammalian Cardiopulmonary Systems. Physiology (Bethesda) 2021; 36:272-291. [PMID: 34431415 DOI: 10.1152/physiol.00004.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The halogens chlorine (Cl2) and bromine (Br2) are highly reactive oxidizing elements with widespread industrial applications and a history of development and use as chemical weapons. When inhaled, depending on the dose and duration of exposure, they cause acute and chronic injury to both the lungs and systemic organs that may result in the development of chronic changes (such as fibrosis) and death from cardiopulmonary failure. A number of conditions, such as viral infections, coexposure to other toxic gases, and pregnancy increase susceptibility to halogens significantly. Herein we review their danger to public health, their mechanisms of action, and the development of pharmacological agents that when administered post-exposure decrease morbidity and mortality.
Collapse
Affiliation(s)
- Dylan R Addis
- Department of Anesthesiology and Perioperative Medicine, Division of Cardiothoracic Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Pediatrics, Division of Neonatology, Children's Hospital, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, Alabama.,Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
12
|
Siraki AG. The many roles of myeloperoxidase: From inflammation and immunity to biomarkers, drug metabolism and drug discovery. Redox Biol 2021; 46:102109. [PMID: 34455146 PMCID: PMC8403760 DOI: 10.1016/j.redox.2021.102109] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022] Open
Abstract
This review provides a practical guide to myeloperoxidase (MPO) and presents to the reader the diversity of its presence in biology. The review provides a historical background, from peroxidase activity to the discovery of MPO, to its role in disease and drug development. MPO is discussed in terms of its necessity, as specific individuals lack MPO expression. An underlying theme presented throughout brings up the question of the benefit and burden of MPO activity. Enzyme structure is discussed, including accurate masses and glycosylation sites. The catalytic cycle of MPO and its corresponding pathways are presented, with a discussion of the importance of the redox couples of the different states of MPO. Cell lines expressing MPO are discussed and practically summarized for the reader, and locations of MPO (primary and secondary) are provided. Useful methods of MPO detection are discussed, and how these can be used for studying disease processes are implied through the presentation of MPO as a biomarker. The presence of MPO in neutrophil extracellular traps is presented, and the activators of the former are provided. Lastly, the transition from drug metabolism to a target for drug development is where the review concludes.
Collapse
Affiliation(s)
- Arno G Siraki
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
13
|
Bittencourt-Mernak MI, Pinheiro NM, da Silva RC, Ponci V, Banzato R, Pinheiro AJMCR, Olivo CR, Tibério IFLC, Lima Neto LG, Santana FPR, Lago JHG, Prado CM. Effects of Eugenol and Dehydrodieugenol B from Nectandra leucantha against Lipopolysaccharide (LPS)-Induced Experimental Acute Lung Inflammation. JOURNAL OF NATURAL PRODUCTS 2021; 84:2282-2294. [PMID: 34264084 DOI: 10.1021/acs.jnatprod.1c00386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Acute lung injury (ALI) is an important public health problem. The present work investigated whether dehydrodieugenol B treatment, a compound isolated from Brazilian plant Nectandra leucantha (Lauraceae), modulates experimental ALI and compared the observed effects to eugenol. Effects of dehydrodieugenol B in vitro in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells were evaluated. The lung and systemic inflammatory profile, lung function, and possible mechanisms involved in BALB/C male mice (6-8 weeks) with ALI induced by LPS instillation (5 mg/kg) was assayed. Dehydrodieugenol B did not affect the cell viability and inhibited the increase in NO release and IL-1β and IL-6 gene expression induced by LPS. In vivo, both compounds reduced lung edema, inflammatory cells, and the IL-6 and IL-1 β levels in bronchoalveolar lavage fluid, as well as reduced inflammatory cell infiltration and those positive to iNOS, MMP-9, and TIMP-1, and reduced the collagen content and the 8-isoprostane expression in lung tissue. Eugenol and dehydrodieugenol B also inhibited the phosphorylation of Jc-Jun-NH2 terminal Kinase (JNK), a signaling protein involved in the MAPKinase pathway. There was no effect of these compounds in lung function. Therefore, eugenol and dehydrodieugenol B ameliorates several features of experimental ALI and could be considered as a pharmacological tool to ameliorate acute lung inflammation.
Collapse
Affiliation(s)
| | - Nathalia M Pinheiro
- Department of Bioscience, Federal University of São Paulo, Santos, SP, 11015-020, Brazil
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Rafael C da Silva
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
| | - Vitor Ponci
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
| | - Rosana Banzato
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Aruanã J M C R Pinheiro
- Universidade CEUMA, São Luís, MA, 65075-120, Brazil
- Programa de Pós-Graduação da Rede BIONORTE, São Luís, MA, 65055-310, Brazil
| | - Clarice R Olivo
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Iolanda F L C Tibério
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Lídio G Lima Neto
- Universidade CEUMA, São Luís, MA, 65075-120, Brazil
- Programa de Pós-Graduação da Rede BIONORTE, São Luís, MA, 65055-310, Brazil
| | - Fernanda P R Santana
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
- Department of Medicine-Nephrology, Federal University of São Paulo, São Paulo, SP, 04023-062, Brazil
| | - João H G Lago
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo André, SP, 09210-170, Brazil
| | - Carla M Prado
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
- Department of Bioscience, Federal University of São Paulo, Santos, SP, 11015-020, Brazil
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| |
Collapse
|
14
|
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: 89] [Impact Index Per Article: 22.3] [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
|
15
|
Amunugama K, Kolar GR, Ford DA. Neutrophil Myeloperoxidase Derived Chlorolipid Production During Bacteria Exposure. Front Immunol 2021; 12:701227. [PMID: 34489949 PMCID: PMC8416994 DOI: 10.3389/fimmu.2021.701227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Neutrophils are the most abundant white blood cells recruited to the sites of infection and inflammation. During neutrophil activation, myeloperoxidase (MPO) is released and converts hydrogen peroxide to hypochlorous acid (HOCl). HOCl reacts with plasmalogen phospholipids to liberate 2-chlorofatty aldehyde (2-ClFALD), which is metabolized to 2-chlorofatty acid (2-ClFA). 2-ClFA and 2-ClFALD are linked with inflammatory diseases and induce endothelial dysfunction, neutrophil extracellular trap formation (NETosis) and neutrophil chemotaxis. Here we examine the neutrophil-derived chlorolipid production in the presence of pathogenic E. coli strain CFT073 and non-pathogenic E. coli strain JM109. Neutrophils cocultured with CFT073 E. coli strain and JM109 E. coli strain resulted in 2-ClFALD production. 2-ClFA was elevated only in CFT073 coculture. NETosis is more prevalent in CFT073 cocultures with neutrophils compared to JM109 cocultures. 2-ClFA and 2-ClFALD were both shown to have significant bactericidal activity, which is more severe in JM109 E. coli. 2-ClFALD metabolic capacity was 1000-fold greater in neutrophils compared to either strain of E. coli. MPO inhibition reduced chlorolipid production as well as bacterial killing capacity. These findings indicate the chlorolipid profile is different in response to these two different strains of E. coli bacteria.
Collapse
Affiliation(s)
- Kaushalya Amunugama
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Grant R. Kolar
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO, United States
- Research Microscopy and Histology Core, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - David A. Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
16
|
Lindén P, Jonasson S, Hemström P, Ålander L, Larsson A, Ågren L, Elfsmark L, Åstot C. Nasal Lavage Fluid as a Biomedical Sample for Verification of Chlorine Exposure. J Anal Toxicol 2021; 46:559-566. [PMID: 34114620 DOI: 10.1093/jat/bkab069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
Chlorine is a toxic chemical that has been used as a chemical warfare agent in recent armed conflicts. There is an urgent need for methods to verify alleged uses of chlorine, and phospholipid chlorohydrins (PL-HOCl) derived from the pulmonary surfactant of exposed victims have previously been proposed as biomarkers of chlorine exposure. Here we describe an improved protocol for the chemical analysis of these biomarkers and its applicability to biomedical samples from chlorine-exposed animals. By the use of a polymeric solid phase-supported transesterification of PL-HOCl using ethanolamine, a common biomarker; oleoyl ethanolamide chlorohydrin (OEA-HOCl), was derived from all the diverse oleoyl PL-HOCl that may be formed by chlorine exposure. Compared to native lipid biomarkers, OEA-HOCl represents a larger biomarker pool and is better suited for nano-liquid chromatography tandem mass spectrometry (nLC-MS/MS analysis), generating 3 amol LOD and a reduced sample carry-over. With the improved protocol, significantly elevated levels of OEA-HOCl was identified in broncho-alveolar lavage fluid (BALF) of chlorine exposed rats, 2-48 hours after exposure. The difficulty of BALF sampling from humans limits the methods usefulness as a verification tool of chlorine exposure. Conversely, nasal lavage fluid (NLF) is readily collected without advanced equipment. In NLF from chlorine-exposed rats, PL-HOCl were identified and significantly elevated levels of the OEA-HOCl biomarker was detected 2- 24 hours after exposure. In order to test the potential of NLF as a biomedical sample for verification of human exposure to chlorine, in-vitro chlorination of human NLF samples was performed. All human in-vitro chlorinated NLF samples exhibited elevated OEA-HOCl biomarker levels, following sample derivatization. This data indicates the potential of human NLF as a biomedical sample for the verification of chlorine exposure but further work is required to develop and validate the method for the use on real-world samples.
Collapse
Affiliation(s)
- Pernilla Lindén
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Sofia Jonasson
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Petrus Hemström
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Lovisa Ålander
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Andreas Larsson
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Lina Ågren
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Linda Elfsmark
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| | - Crister Åstot
- The Swedish Defence Research Agency, CBRN Defence and Security, Cementvägen 20, Umeå, Sweden
| |
Collapse
|
17
|
Amunugama K, Pike DP, Ford DA. The lipid biology of sepsis. J Lipid Res 2021; 62:100090. [PMID: 34087197 PMCID: PMC8243525 DOI: 10.1016/j.jlr.2021.100090] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/12/2023] Open
Abstract
Sepsis, defined as the dysregulated immune response to an infection leading to organ dysfunction, is one of the leading causes of mortality around the globe. Despite the significant progress in delineating the underlying mechanisms of sepsis pathogenesis, there are currently no effective treatments or specific diagnostic biomarkers in the clinical setting. The perturbation of cell signaling mechanisms, inadequate inflammation resolution, and energy imbalance, all of which are altered during sepsis, are also known to lead to defective lipid metabolism. The use of lipids as biomarkers with high specificity and sensitivity may aid in early diagnosis and guide clinical decision making. In addition, identifying the link between specific lipid signatures and their role in sepsis pathology may lead to novel therapeutics. In this review, we discuss the recent evidence on dysregulated lipid metabolism both in experimental and human sepsis focused on bioactive lipids, fatty acids, and cholesterol as well as the enzymes regulating their levels during sepsis. We highlight not only their potential roles in sepsis pathogenesis but also the possibility of using these respective lipid compounds as diagnostic and prognostic biomarkers of sepsis.
Collapse
Affiliation(s)
- Kaushalya Amunugama
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel P Pike
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Prasch J, Bernhart E, Reicher H, Kollroser M, Rechberger GN, Koyani CN, Trummer C, Rech L, Rainer PP, Hammer A, Malle E, Sattler W. Myeloperoxidase-Derived 2-Chlorohexadecanal Is Generated in Mouse Heart during Endotoxemia and Induces Modification of Distinct Cardiomyocyte Protein Subsets In Vitro. Int J Mol Sci 2020; 21:ijms21239235. [PMID: 33287422 PMCID: PMC7730634 DOI: 10.3390/ijms21239235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Sepsis is a major cause of mortality in critically ill patients and associated with cardiac dysfunction, a complication linked to immunological and metabolic aberrations. Cardiac neutrophil infiltration and subsequent release of myeloperoxidase (MPO) leads to the formation of the oxidant hypochlorous acid (HOCl) that is able to chemically modify plasmalogens (ether-phospholipids) abundantly present in the heart. This reaction gives rise to the formation of reactive lipid species including aldehydes and chlorinated fatty acids. During the present study, we tested whether endotoxemia increases MPO-dependent lipid oxidation/modification in the mouse heart. In hearts of lipopolysaccharide-injected mice, we observed significantly higher infiltration of MPO-positive cells, increased fatty acid content, and formation of 2-chlorohexadecanal (2-ClHDA), an MPO-derived plasmalogen modification product. Using murine HL-1 cardiomyocytes as in vitro model, we show that exogenously added HOCl attacks the cellular plasmalogen pool and gives rise to the formation of 2-ClHDA. Addition of 2-ClHDA to HL-1 cardiomyocytes resulted in conversion to 2-chlorohexadecanoic acid and 2-chlorohexadecanol, indicating fatty aldehyde dehydrogenase-mediated redox metabolism. However, a recovery of only 40% indicated the formation of non-extractable (protein) adducts. To identify protein targets, we used a clickable alkynyl analog, 2-chlorohexadec-15-yn-1-al (2-ClHDyA). After Huisgen 1,3-dipolar cycloaddition of 5-tetramethylrhodamine azide (N3-TAMRA) and two dimensional-gel electrophoresis (2D-GE), we were able to identify 51 proteins that form adducts with 2-ClHDyA. Gene ontology enrichment analyses revealed an overrepresentation of heat shock and chaperone, energy metabolism, and cytoskeletal proteins as major targets. Our observations in a murine endotoxemia model demonstrate formation of HOCl-modified lipids in the heart, while pathway analysis in vitro revealed that the chlorinated aldehyde targets specific protein subsets, which are central to cardiac function.
Collapse
Affiliation(s)
- Jürgen Prasch
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Eva Bernhart
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | | | - Gerald N. Rechberger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria;
- Center for Explorative Lipidomics, BioTechMed Graz, 8010 Graz, Austria
| | - Chintan N. Koyani
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Christopher Trummer
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Lavinia Rech
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Peter P. Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Astrid Hammer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, 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; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Wolfgang Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
- Center for Explorative Lipidomics, BioTechMed Graz, 8010 Graz, Austria
- Correspondence: ; Tel.: +43-316-385-71950
| |
Collapse
|
19
|
Wu X, Lin L, Wu H. Ferulic acid alleviates lipopolysaccharide-induced acute lung injury through inhibiting TLR4/NF-κB signaling pathway. J Biochem Mol Toxicol 2020; 35:e22664. [PMID: 33140555 DOI: 10.1002/jbt.22664] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/15/2020] [Accepted: 10/20/2020] [Indexed: 01/05/2023]
Abstract
Ferulic acid (FA) exhibits anti-inflammatory, antidiabetic, antihyperlipidemic, antioxidant, neuroprotective, and antihypertensive effects. This study aimed to determine whether FA could ameliorate lipopolysaccharide (LPS)-induced inflammatory responses and acute lung injury (ALI) in mice. Mice were challenged with LPS intratracheally to induce ALI 1 h after 3 days of FA (25, 50, and 100 mg/kg) or dexamethasone (DEX; 5 mg/kg) administration. The lung tissues and bronchoalveolar lavage fluid (BALF) were collected 12 h after the LPS challenge. Pretreatment with FA or DEX could attenuate lung histopathological change, complement deposition, and lung wet-to-dry weight ratio of mice injured by LPS. Meanwhile, the influx of neutrophils and macrophages, as well as the production of proinflammatory cytokine (tumor necrosis factor-alpha, interleukin 1 beta [IL-1β], and IL-6), in BALF of ALI mice was significantly decreased. Moreover, FA or DEX markedly reversed the LPS-induced elevation of myeloperoxidase activity and monocyte chemoattractant protein-1 level in lung tissues of ALI mice. In addition, the Western blot analysis demonstrated that FA or DEX effectively inhibited the LPS-induced activation of the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway in lung tissues. The current study suggested that the alleviating effect of FA against LPS-induced ALI might be partially due to the inhibition of the inflammatory response via inactivation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xialei Wu
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Liyao Lin
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Haibin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
20
|
Lazrak A, Song W, Zhou T, Aggarwal S, Jilling T, Garantziotis S, Matalon S. Hyaluronan and halogen-induced airway hyperresponsiveness and lung injury. Ann N Y Acad Sci 2020; 1479:29-43. [PMID: 32578230 PMCID: PMC7680259 DOI: 10.1111/nyas.14415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
Chlorine (Cl2 ) and bromine (Br2 ) are produced in large quantities throughout the world and used in the industry and the sanitation of water. These halogens can pose a significant threat to public health when released into the atmosphere during transportation and industrial accidents, or as acts of terrorism. In this review, we discuss the evidence showing that the activity of Cl2 and Br2 , and of products formed by their interaction with biomolecules, fragment high-molecular-weight hyaluronan (HMW-HA), a key component of the interstitial space and present in epithelial cells, to form proinflammatory, low-molecular-weight hyaluronan fragments that increase intracellular calcium (Ca2+ ) and activate RAS homolog family member A (RhoA) in airway smooth muscle and epithelial and microvascular cells. These changes result in airway hyperresponsiveness (AHR) to methacholine and increase epithelial and microvascular permeability. The increase in intracellular Ca2+ is the result of the activation of the calcium-sensing receptor by Cl2 , Br2 , and their by-products. Posthalogen administration of a commercially available form of HMW-HA to mice and to airway cells in vitro reverses the increase of Ca2+ and the activation of RhoA, and restores AHR to near-normal levels of airway function. These data have established the potential of HMW-HA to be a countermeasure against Cl2 and Br2 toxicity.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Division of Molecular and Translational Biomedicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Weifeng Song
- Division of Molecular and Translational Biomedicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Ting Zhou
- Division of Molecular and Translational Biomedicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Tamas Jilling
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Division of Neonatology, Department of Pediatrics, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Stavros Garantziotis
- Matrix Biology Group, Immunity, Inflammation, and Disease Laboratory, NIH/NIEHS, RTP, NC
| | - Sadis Matalon
- Division of Molecular and Translational Biomedicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
- Pulmonary Injury and Repair Center, Department of Anesthesiology and Perioperative Medicine, the University of Alabama at Birmingham School of Medicine, Birmingham, AL
| |
Collapse
|
21
|
Myeloperoxidase: A versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther 2020; 221:107711. [PMID: 33137376 DOI: 10.1016/j.pharmthera.2020.107711] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Myeloperoxidase (MPO) is a prominent mammalian heme peroxidase and a fundamental component of the innate immune response against microbial pathogens. In recent times, MPO has received considerable attention as a key oxidative enzyme capable of impairing the bioactivity of nitric oxide (NO) and promoting endothelial dysfunction; a clinically relevant event that manifests throughout the development of inflammatory cardiovascular disease. Increasing evidence indicates that during cardiovascular disease, MPO is released intravascularly by activated leukocytes resulting in its transport and sequestration within the vascular endothelium. At this site, MPO catalyzes various oxidative reactions that are capable of promoting vascular inflammation and impairing NO bioactivity and endothelial function. In particular, MPO catalyzes the production of the potent oxidant hypochlorous acid (HOCl) and the catalytic consumption of NO via the enzyme's NO oxidase activity. An emerging paradigm is the ability of MPO to also influence endothelial function via non-catalytic, cytokine-like activities. In this review article we discuss the implications of our increasing knowledge of the versatility of MPO's actions as a mediator of cardiovascular disease and endothelial dysfunction for the development of new pharmacological agents capable of effectively combating MPO's pathogenic activities. More specifically, we will (i) discuss the various transport mechanisms by which MPO accumulates into the endothelium of inflamed or diseased arteries, (ii) detail the clinical and basic scientific evidence identifying MPO as a significant cause of endothelial dysfunction and cardiovascular disease, (iii) provide an up-to-date coverage on the different oxidative mechanisms by which MPO can impair endothelial function during cardiovascular disease including an evaluation of the contributions of MPO-catalyzed HOCl production and NO oxidation, and (iv) outline the novel non-enzymatic mechanisms of MPO and their potential contribution to endothelial dysfunction. Finally, we deliver a detailed appraisal of the different pharmacological strategies available for targeting the catalytic and non-catalytic modes-of-action of MPO in order to protect against endothelial dysfunction in cardiovascular disease.
Collapse
|
22
|
Yu H, Liu Y, Wang M, Restrepo RJ, Wang D, Kalogeris TJ, Neumann WL, Ford DA, Korthuis RJ. Myeloperoxidase instigates proinflammatory responses in a cecal ligation and puncture rat model of sepsis. Am J Physiol Heart Circ Physiol 2020; 319:H705-H721. [PMID: 32762560 DOI: 10.1152/ajpheart.00440.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloperoxidase (MPO)-derived hypochlorous (HOCl) reacts with membrane plasmalogens to yield α-chlorofatty aldehydes such as 2-chlorofatty aldehyde (2-ClFALD) and its metabolite 2-chlorofatty acid (2-ClFA). Recent studies showed that 2-ClFALD and 2-ClFA serve as mediators of the inflammatory responses to sepsis by as yet unknown mechanisms. Since no scavenger for chlorinated lipids is available and on the basis of the well-established role of the MPO/HOCl/chlorinated lipid axis in inflammatory responses, we hypothesized that treatment with MPO inhibitors (N-acetyl lysyltyrosylcysteine amide or 4-aminobenzoic acid hydrazide) would inhibit inflammation and proinflammatory mediator expression induced by cecal ligation and puncture (CLP). We used intravital microscopy to quantify in vivo inflammatory responses in Sham and CLP rats with or without MPO inhibition. Small intestines, mesenteries, and lungs were collected to assess changes in MPO-positive staining and lung injury, respectively, as well as free 2-ClFA and proinflammatory mediators levels. CLP caused neutrophil infiltration, 2-ClFA generation, acute lung injury, leukocyte-/platelet-endothelium interactions, mast cell activation (MCA), plasminogen activator inhibitor-1 (PAI-1) production, and the expression of several cytokines, chemokines, and vascular endothelial growth factor, changes that were reduced by MPO inhibition. Pretreatment with a PAI-1 inhibitor or MC stabilizer prevented CLP-induced leukocyte-endothelium interactions and MCA, and abrogated exogenous 2-ClFALD-induced inflammatory responses. Thus, we provide evidence that MPO instigates these inflammatory changes in CLP and that chlorinated lipids may serve as a mechanistic link between the enzymatic activity of MPO and PAI-1- and mast cell-dependent adhesive interactions, providing a rationale for new therapeutic interventions in sepsis.NEW & NOTEWORTHY Using two distinct myeloperoxidase (MPO) inhibitors, we show for the first time that MPO plays an important role in producing increases in free 2-chlorofatty aldehyde (2-ClFALD)-a powerful proinflammatory chlorinated lipid in plasma and intestine-a number of cytokines and other inflammatory mediators, leukocyte and platelet rolling and adhesion in postcapillary venules, and lung injury in a cecal ligation and puncture model of sepsis. In addition, the use of a plasminogen activator inhibitor-1 (PAI-1) inhibitor or a mast cell stabilizer prevented inflammatory responses in CLP-induced sepsis. PAI-1 inhibition also prevented the proinflammatory responses to exogenous 2-ClFALD superfusion. Thus, our study provides some of the first evidence that MPO-derived free 2-ClFA plays an important role in CLP-induced sepsis by a PAI-1- and mast cell-dependent mechanism.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Yajun Liu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Ricardo J Restrepo
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Derek Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Theodore J Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - William L Neumann
- Department of Pharmaceutical Sciences, Edwardsville School of Pharmacy, Southern Illinois University, Edwardsville, Illinois
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Center for Cardiovascular Research, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
23
|
Ramirez DC, Gomez Mejiba SE. Pulmonary Neutrophilic Inflammation and Noncommunicable Diseases: Pathophysiology, Redox Mechanisms, Biomarkers, and Therapeutics. Antioxid Redox Signal 2020; 33:211-227. [PMID: 32319787 DOI: 10.1089/ars.2020.8098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Pulmonary neurophilic inflammation (PNI) is the homing and activation of neutrophil with damage to the microvasculature. This process is involved in pulmonary damage in patients exposed to airborne pollutants (exogenous stressors) and also to systemic inflammation/oxidative stress (endogenous stressors) associated with noncommunicable diseases (NCDs). Recent Advances: PNI is an important trigger of the early onset and progression of NCD in susceptible patients exposed to airborne pollutants. Irritation of the lung microvasculature by exogenous and endogenous stressors causes PNI. Circulating endogenous stressors in NCD can cause PNI. Critical Issues: Air pollution-triggered PNI causes increased circulating endogenous stressors that can trigger NCD in susceptible patients. Systemic inflammation/oxidative stress associated with NCD can cause PNI. Inflammation/end-oxidation products of macromolecules are also potential biomarkers and therapeutic targets for NCD-triggered PNI- and PNI-triggered NCD. Future Directions: Understanding the molecular mechanism of PNI triggered by exogenous or endogenous stressors will help explain the early onset of NCD in susceptible patients exposed to air pollution. It can also help undercover biomarkers and mechanism-based therapeutic targets in air pollutant-triggered PNI, PNI-triggered NCD, and NCD-triggered PNI.
Collapse
Affiliation(s)
- Dario C Ramirez
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL, CCT-San Luis, CONICET, School of Chemistry, Biochemistry and Pharmacy, National University of San Luis, San Luis, Argentina
| | - Sandra E Gomez Mejiba
- Laboratory of Experimental Therapeutics and Nutrition, IMIBIO-SL, CCT-San Luis, CONICET, School of Health Sciences, National University of San Luis, San Luis, Argentina
| |
Collapse
|
24
|
Reactive species generated by heme impair alveolar epithelial sodium channel function in acute respiratory distress syndrome. Redox Biol 2020; 36:101592. [PMID: 32506040 PMCID: PMC7276446 DOI: 10.1016/j.redox.2020.101592] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
We previously reported that the highly reactive cell-free heme (CFH) is increased in the plasma of patients with chronic lung injury and causes pulmonary edema in animal model of acute respiratory distress syndrome (ARDS) post inhalation of halogen gas. However, the mechanisms by which CFH causes pulmonary edema are unclear. Herein we report for the first time that CFH and chlorinated lipids (formed by the interaction of halogen gas, Cl2, with plasmalogens) are increased in the plasma of patients exposed to Cl2 gas. Ex vivo incubation of red blood cells (RBC) with halogenated lipids caused oxidative damage to RBC cytoskeletal protein spectrin, resulting in hemolysis and release of CFH. Patch clamp and short circuit current measurements revealed that CFH inhibited the activity of amiloride-sensitive epithelial Na+ channel (ENaC) and cation sodium (Na+) channels in mouse alveolar cells and trans-epithelial Na+ transport across human airway cells with EC50 of 125 nM and 500 nM, respectively. Molecular modeling identified 22 putative heme-docking sites on ENaC (energy of binding range: 86-1563 kJ/mol) with at least 2 sites within its narrow transmembrane pore, potentially capable of blocking Na+ transport across the channel. A single intramuscular injection of the heme-scavenging protein, hemopexin (4 μg/kg body weight), one hour post halogen gas exposure, decreased plasma CFH and improved lung ENaC activity in mice. In conclusion, results suggested that CFH mediated inhibition of ENaC activity may be responsible for pulmonary edema post inhalation injury.
Collapse
|
25
|
van der Zee P, Rietdijk W, Somhorst P, Endeman H, Gommers D. A systematic review of biomarkers multivariately associated with acute respiratory distress syndrome development and mortality. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:243. [PMID: 32448370 PMCID: PMC7245629 DOI: 10.1186/s13054-020-02913-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Abstract
Background Heterogeneity of acute respiratory distress syndrome (ARDS) could be reduced by identification of biomarker-based phenotypes. The set of ARDS biomarkers to prospectively define these phenotypes remains to be established. Objective To provide an overview of the biomarkers that were multivariately associated with ARDS development or mortality. Data sources We performed a systematic search in Embase, MEDLINE, Web of Science, Cochrane CENTRAL, and Google Scholar from inception until 6 March 2020. Study selection Studies assessing biomarkers for ARDS development in critically ill patients at risk for ARDS and mortality due to ARDS adjusted in multivariate analyses were included. Data extraction and synthesis We included 35 studies for ARDS development (10,667 patients at risk for ARDS) and 53 for ARDS mortality (15,344 patients with ARDS). These studies were too heterogeneous to be used in a meta-analysis, as time until outcome and the variables used in the multivariate analyses varied widely between studies. After qualitative inspection, high plasma levels of angiopoeitin-2 and receptor for advanced glycation end products (RAGE) were associated with an increased risk of ARDS development. None of the biomarkers (plasma angiopoeitin-2, C-reactive protein, interleukin-8, RAGE, surfactant protein D, and Von Willebrand factor) was clearly associated with mortality. Conclusions Biomarker data reporting and variables used in multivariate analyses differed greatly between studies. Angiopoeitin-2 and RAGE in plasma were positively associated with increased risk of ARDS development. None of the biomarkers independently predicted mortality. Therefore, we suggested to structurally investigate a combination of biomarkers and clinical parameters in order to find more homogeneous ARDS phenotypes. PROSPERO identifier PROSPERO, CRD42017078957
Collapse
Affiliation(s)
- Philip van der Zee
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - Wim Rietdijk
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Peter Somhorst
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Henrik Endeman
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Diederik Gommers
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Carr AC, Spencer E, Hoskin TS, Rosengrave P, Kettle AJ, Shaw G. Circulating myeloperoxidase is elevated in septic shock and is associated with systemic organ failure and mortality in critically ill patients. Free Radic Biol Med 2020; 152:462-468. [PMID: 31698081 DOI: 10.1016/j.freeradbiomed.2019.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/29/2019] [Accepted: 11/03/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Neutrophils are elevated in critically ill patients during the systemic inflammatory response to trauma and sepsis. The neutrophil-derived enzyme myeloperoxidase generates reactive oxygen species which can react with host tissue resulting in cell damage and dysfunction. Thus, elevated myeloperoxidase in the circulation may be associated with adverse patient outcomes. METHODS Circulating myeloperoxidase concentrations were measured in a cohort of 44 critically ill patients, 55% of whom were diagnosed with septic shock, and 44 healthy controls. Intensive care mortality prediction scores (SOFA, SAPS, APACHE) and ICU and hospital mortality were obtained from the patients' clinical notes. Hematological and biochemical assessments included blood cell counts, lactate, alanine transaminase, creatinine, bilirubin, C-reactive protein, and PaO2. Myeloperoxidase was measured using a commercial ELISA kit and cell free DNA was detected using SytoxGreen™ fluorescence staining. RESULTS Myeloperoxidase concentrations were significantly higher in critically ill patients than control samples (234 ± 30 ng/ml versus 15 ± 4 ng/ml, p < 0.001), and were elevated in septic shock relative to non-septic patients (302 ± 42 ng/ml versus 156 ± 38 ng/ml, p = 0.02), despite neutrophil counts being comparable between the two subgroups (p = 0.6). Myeloperoxidase correlated with SOFA scores in the critically ill patients (r = 0.395, p = 0.02), and with markers of tissue dysfunction and injury such as lactate (r = 0.572, p < 0.001), log10 alanine transferase (r = 0.392, p = 0.016) and log10 cell free DNA (r = 0.371, p = 0.03). The subgroup of patients with higher than mean APACHE III scores (i.e. >78, n = 16) exhibited significantly elevated myeloperoxidase concentrations in the non-survivors compared with survivors (416 ± 59 ng/ml versus 140 ± 33 ng/mL, p = 0.001). Hospital mortality for the whole cohort was 27%; mortality in the high APACHE III subgroup was 38%, and when combined with higher than mean myeloperoxidase (i.e. >234 ng/mL), mortality increased to 71%. CONCLUSIONS Myeloperoxidase is associated with markers of tissue injury and systemic organ failure, particularly in septic patients. The enzyme is also associated with mortality in patients with higher APACHE III scores, and thus has potential as an additional diagnostic marker to improve mortality prediction.
Collapse
Affiliation(s)
- Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| | - Emma Spencer
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Teagan S Hoskin
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Patrice Rosengrave
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Geoffrey Shaw
- Department of Intensive Care, Christchurch Hospital, Christchurch, New Zealand
| |
Collapse
|
27
|
McHowat J, Shakya S, Ford DA. 2-Chlorofatty Aldehyde Elicits Endothelial Cell Activation. Front Physiol 2020; 11:460. [PMID: 32457656 PMCID: PMC7225355 DOI: 10.3389/fphys.2020.00460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Endothelial activation and dysfunction are hallmarks of inflammation. Neutrophil-vascular endothelium interactions have significant effects on vascular wall physiology and pathology. Myeloperoxidase (MPO)-derived products released from activated neutrophils can mediate the inflammatory response and contribute to endothelial dysfunction. 2-Chlorofatty aldehyde (2-ClFALD) is the direct oxidation product of MPO-derived hypochlorous acid (HOCl) targeting plasmalogen phospholipids. The role of 2-ClFALD in endothelial dysfunction is poorly understood and may be dependent on the vascular bed. This study compared the role of 2-ClFALD in eliciting endothelial dysfunction in human coronary artery endothelial cells (HCAEC), human lung microvascular endothelial cells (HLMVEC), and human kidney endothelial cells (HKEC). Profound increases in selectin surface expression as well as ICAM-1 and VCAM-1 surface expression were observed in HCAEC and HLMVEC. The surface expression of these adherence molecules resulted in robust adherence of neutrophils and platelets to 2-ClFALD treated endothelial cells. In contrast to HCAEC and HLMVEC, 2-ClFALD-treated HKEC had substantially reduced adherence molecule surface expression with no resulting increase in platelet adherence. 2-ClFALD-treated HKEC did have an increase in neutrophil adherence. All three endothelial cell lines treated with 2-ClFALD displayed a time-dependent loss of barrier function. Further studies revealed 2-ClHDyA localizes to ER and Golgi when using a synthetic alkyne analog of 2-ClFALD in HCAEC and HLMVEC. These findings indicate 2-ClFALDs promote endothelial cell dysfunction with disparate degrees of responsiveness depending on the vascular bed of origin.
Collapse
Affiliation(s)
- Jane McHowat
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO, United States.,Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Shubha Shakya
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States.,Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - David A Ford
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States.,Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
28
|
Pike DP, Vogel MJ, McHowat J, Mikuzis PA, Schulte KA, Ford DA. 2-Chlorofatty acids are biomarkers of sepsis mortality and mediators of barrier dysfunction in rats. J Lipid Res 2020; 61:1115-1127. [PMID: 32376642 DOI: 10.1194/jlr.ra120000829] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is defined as the systemic, dysregulated host immune response to an infection that leads to injury to host organ systems and, often, death. Complex interactions between pathogens and their hosts elicit microcirculatory dysfunction. Neutrophil myeloperoxidase (MPO) is critical for combating pathogens, but MPO-derived hypochlorous acid (HOCl) can react with host molecular species as well. Plasmalogens are targeted by HOCl, leading to the production of 2-chlorofatty acids (2-CLFAs). 2-CLFAs are associated with human sepsis mortality, decrease in vitro endothelial barrier function, and activate human neutrophil extracellular trap formation. Here, we sought to examine 2-CLFAs in an in vivo rat sepsis model. Intraperitoneal cecal slurry sepsis with clinically relevant rescue therapies led to ∼73% mortality and evidence of microcirculatory dysfunction. Plasma concentrations of 2-CLFAs assessed 8 h after sepsis induction were lower in rats that survived sepsis than in nonsurvivors. 2-CLFA levels were elevated in kidney, liver, spleen, lung, colon, and ileum in septic animals. In vivo, exogenous 2-CLFA treatments increased kidney permeability, and in in vitro experiments, 2-CLFA also increased epithelial surface expression of vascular cell adhesion molecule 1 and decreased epithelial barrier function. Collectively, these studies support a role of free 2-CLFAs as biomarkers of sepsis mortality, potentially mediated, in part, by 2-CLFA-elicited endothelial and epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Daniel P Pike
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Michael J Vogel
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Jane McHowat
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104; Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Paul A Mikuzis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Kevin A Schulte
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104. mailto:
| |
Collapse
|
29
|
Abstract
Increased endothelial cell adhesion molecule (ECAM) expression, leukocyte-endothelial cell adhesive interactions (LECA), platelet-endothelial cell adhesion (PECA), mast cell activation, production of reactive oxygen species (ROS), and microvascular permeability are hallmarks of the inflammatory response. The infiltration of inflammatory phagocytes is associated with myeloperoxidase (MPO)-dependent production of hypochlorous acid, a reactive chlorinating species that targets membrane lipids to produce halogenated lipids such as 2-chlorohexadecanal (2-ClHDA) and 2-chloropalmitic acid (2-ClPA). Whether these chlorinated lipids contribute to microcirculatory dysfunction is largely unknown. Thus, the objectives of this study were to determine if chlorinated lipids exposure induces such inflammatory responses in an in vitro model employing cultured human intestinal mesenteric vascular endothelial cells (HIMVEC), and in an in vivo model examining responses in small intestinal and mesenteric postcapillary venules of naive rats. Following the addition of either 2-ClPA or 2-ClHDA to the culture medium, HIMVEC displayed increased platelet and neutrophil adherence that was associated with elevated expression of ECAMs and increased permeability. In vivo, chlorinated lipid exposure significantly increased LECA, PECA, ROS production, and albumin leakage, inflammatory events that were associated with mast cell activation and increased tissue MPO activity and expression. Our data provide proof-of-principle that 2-ClPA and 2-ClHDA induce powerful proinflammatory responses both in vitro and in vivo, suggesting the possibility that these chlorinated lipid products of the MPO/ hydrogen peroxide /chloride system may contribute to inflammation noted in neutrophil-dependent, myeloperoxidase-mediated pathologic states such as ischemia/reperfusion, hemorrhagic shock, and sepsis.
Collapse
|
30
|
Goeritzer M, Bernhart E, Plastira I, Reicher H, Leopold C, Eichmann TO, Rechberger G, Madreiter-Sokolowski CT, Prasch J, Eller P, Graier WF, Kratky D, Malle E, Sattler W. Myeloperoxidase and Septic Conditions Disrupt Sphingolipid Homeostasis in Murine Brain Capillaries In Vivo and Immortalized Human Brain Endothelial Cells In Vitro. Int J Mol Sci 2020; 21:E1143. [PMID: 32050431 PMCID: PMC7037060 DOI: 10.3390/ijms21031143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
During inflammation, activated leukocytes release cytotoxic mediators that compromise blood-brain barrier (BBB) function. Under inflammatory conditions, myeloperoxidase (MPO) is critically involved in inflicting BBB damage. We used genetic and pharmacological approaches to investigate whether MPO induces aberrant lipid homeostasis at the BBB in a murine endotoxemia model. To corroborate findings in a human system we studied the impact of sera from sepsis and non-sepsis patients on brain endothelial cells (hCMEC/D3). In response to endotoxin, the fatty acid, ceramide, and sphingomyelin content of isolated mouse brain capillaries dropped and barrier dysfunction occurred. In mice, genetic deficiency or pharmacological inhibition of MPO abolished these alterations. Studies in metabolic cages revealed increased physical activity and less pronounced sickness behavior of MPO-/- compared to wild-type mice in response to sepsis. In hCMEC/D3 cells, exogenous tumor necrosis factor α (TNFα) potently regulated gene expression of pro-inflammatory cytokines and a set of genes involved in sphingolipid (SL) homeostasis. Notably, treatment of hCMEC/D3 cells with sera from septic patients reduced cellular ceramide concentrations and induced barrier and mitochondrial dysfunction. In summary, our in vivo and in vitro data revealed that inflammatory mediators including MPO, TNFα induce dysfunctional SL homeostasis in brain endothelial cells. Genetic and pharmacological inhibition of MPO attenuated endotoxin-induced alterations in SL homeostasis in vivo, highlighting the potential role of MPO as drug target to treat inflammation-induced brain dysfunction.
Collapse
Affiliation(s)
- Madeleine Goeritzer
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
- BioTechMed-Graz, Graz 8010, Austria; (T.O.E.); (G.R.)
| | - Eva Bernhart
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Ioanna Plastira
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Christina Leopold
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Thomas O. Eichmann
- BioTechMed-Graz, Graz 8010, Austria; (T.O.E.); (G.R.)
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz 8010, Austria
| | - Gerald Rechberger
- BioTechMed-Graz, Graz 8010, Austria; (T.O.E.); (G.R.)
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz 8010, Austria
| | - Corina T. Madreiter-Sokolowski
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
- Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Jürgen Prasch
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Philipp Eller
- Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Graz 8036, Austria;
| | - Wolfgang F. Graier
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Dagmar Kratky
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
- BioTechMed-Graz, Graz 8010, Austria; (T.O.E.); (G.R.)
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
| | - Wolfgang Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz 8010, Austria; (M.G.); (E.B.); (I.P.); (H.R.); (C.L.); (C.T.M.-S.); (J.P.); (W.F.G.); (D.K.); (E.M.)
- BioTechMed-Graz, Graz 8010, Austria; (T.O.E.); (G.R.)
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz 8010, Austria
| |
Collapse
|
31
|
Spickett CM. Formation of Oxidatively Modified Lipids as the Basis for a Cellular Epilipidome. Front Endocrinol (Lausanne) 2020; 11:602771. [PMID: 33408694 PMCID: PMC7779974 DOI: 10.3389/fendo.2020.602771] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
While often regarded as a subset of metabolomics, lipidomics can better be considered as a field in its own right. While the total number of lipid species in biology may not exceed the number of metabolites, they can be modified chemically and biochemically leading to an enormous diversity of derivatives, many of which retain the lipophilic properties of lipids and thus expand the lipidome greatly. Oxidative modification by radical oxygen species, either enzymatically or chemically, is one of the major mechanisms involved, although attack by non-radical oxidants also occurs. The modified lipids typically contain more oxygens in the form of hydroxyl, epoxide, carbonyl and carboxylic acid groups, and nitration, nitrosylation, halogenation or sulfation can also occur. This article provides a succinct overview of the types of species formed, the reactive compounds involved and the specific molecular sites that they react with, and the biochemical or chemical mechanisms involved. In many cases, these modifications reduce the stability of the lipid, and breakdown products are formed, which themselves have interesting properties such as the ability to react with other biomolecules. Publications on the biological effects of modified lipids are growing rapidly, supporting the concept that some of these biomolecules have potential signaling and regulatory effects. The question therefore arises whether modified lipids represent an "epilipidome", analogous to the epigenetic modifications that can control gene expression.
Collapse
|
32
|
Hartman CL, Ford DA. MPO (Myeloperoxidase) Caused Endothelial Dysfunction. Arterioscler Thromb Vasc Biol 2019; 38:1676-1677. [PMID: 30354198 DOI: 10.1161/atvbaha.118.311427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Celine L Hartman
- From the Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, MO
| | - David A Ford
- From the Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, MO
| |
Collapse
|
33
|
Huang X, Zhao M. High expression of long non-coding RNA MALAT1 correlates with raised acute respiratory distress syndrome risk, disease severity, and increased mortality in sepstic patients. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1877-1887. [PMID: 31934011 PMCID: PMC6947113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/25/2019] [Indexed: 06/10/2023]
Abstract
This study aimed to explore the correlation of long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (lnc-MALAT1) expression with acute respiratory distress syndrome (ARDS) risk, disease severity, inflammation level, and mortality in septic patients. 152 septic patients were consecutively included and surveillance was conducted daily to identify ARDS occurrence. Severity and organ failure of sepsis were assessed by APACHE II score and SOFA score respectively, and the in-hospital mortality was calculated. Patients' blood samples were extracted. Lnc-MALAT1 expression and inflammatory cytokines levels were detected using real-time qPCR and ELISA assay respectively. The incidence of ARDS was 27.0%. Lnc-MALAT1 expression was increased in ARDS patients compared to non-ARDS patients, and it could distinguish ARDS from non-ARDS by ROC with AUC of 0.674 (95% CI: 0.581-0.766). Multivariate logistic regression analysis displayed that lnc-MALAT1 high expression, increased age, higher proportion of smoking and COPD were independent factors for predicting elevated ARDS risk. Lnc-MALAT1 expression was positively correlated with APACHE II score, SOFA score, and inflammatory factors levels including C-reactive protein, procalcitonin, TNF-α, IL-1β, IL-6 and IL-17. Furthermore, the mortality was 30.9%, and lnc-MALAT1 expression was elevated in non-survivors compared to survivors, presenting a good predictive value for high mortality by ROC with AUC of 0.651 (95% CI: 0.555-0.747). Lnc-MALAT1 high expression predicts increased ARDS risk, and correlates with severe disease condition and raised mortality in sepsis patients.
Collapse
Affiliation(s)
- Xing Huang
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430014, China
| | - Min Zhao
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430014, China
| |
Collapse
|
34
|
Meng LB, Chen K, Zhang YM, Gong T. Common Injuries and Repair Mechanisms in the Endothelial Lining. Chin Med J (Engl) 2018; 131:2338-2345. [PMID: 30246720 PMCID: PMC6166454 DOI: 10.4103/0366-6999.241805] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective: Endothelial cells (ECs) are important metabolic and endocrinal organs which play a significant role in regulating vascular function. Vascular ECs, located between the blood and vascular tissues, can not only complete the metabolism of blood and interstitial fluid but also synthesize and secrete a variety of biologically active substances to maintain vascular tension and keep a normal flow of blood and long-term patency. Therefore, this article presents a systematic review of common injuries and healing mechanisms for the vascular endothelium. Data Sources: An extensive search in the PubMed database was undertaken, focusing on research published after 2003 with keywords including endothelium, vascular, wounds and injuries, and wound healing. Study Selection: Several types of articles, including original studies and literature reviews, were identified and reviewed to summarize common injury and repair processes of the endothelial lining. Results: Endothelial injury is closely related to the development of multiple cardiovascular and cerebrovascular diseases. However, the mechanism of vascular endothelial injury is not fully understood. Numerous studies have shown that the mechanisms of EC injury mainly involve inflammatory reactions, physical stimulation, chemical poisons, concurrency of related diseases, and molecular changes. Endothelial progenitor cells play an important role during the process of endothelial repair after such injuries. What's more, a variety of restorative cells, changes in cytokines and molecules, chemical drugs, certain RNAs, regulation of blood pressure, and physical fitness training protect the endothelial lining by reducing the inducing factors, inhibiting inflammation and oxidative stress reactions, and delaying endothelial caducity. Conclusions: ECs are always in the process of being damaged. Several therapeutic targets and drugs were seeked to protect the endothelium and promote repair.
Collapse
Affiliation(s)
- Ling-Bing Meng
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Kun Chen
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Yuan-Meng Zhang
- Department of Internal Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Tao Gong
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| |
Collapse
|
35
|
Biosynthesis of human myeloperoxidase. Arch Biochem Biophys 2018; 642:1-9. [PMID: 29408362 DOI: 10.1016/j.abb.2018.02.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 01/30/2023]
Abstract
Members of Chordata peroxidase subfamily [1] expressed in mammals, including myeloperoxidase (MPO), eosinophil peroxidase (EPO), lactoperoxidase (LPO), and thyroid peroxidase (TPO), express conserved motifs around the heme prosthetic group essential for their activity, a calcium-binding site, and at least two covalent bonds linking the heme group to the protein backbone. Although most studies of the biosynthesis of these peroxidases have focused on MPO, many of the features described occur during biosynthesis of other members of the protein subfamily. Whereas MPO biosynthesis includes events typical for proteins generated in the secretory pathway, the importance and consequences of heme insertion are events uniquely associated with peroxidases. This Review summarizes decades of work elucidating specific steps in the biosynthetic pathway of human MPO. Discussion includes cotranslational glycosylation and subsequent modifications of the N-linked carbohydrate sidechains, contributions by molecular chaperones in the endoplasmic reticulum, cleavage of the propeptide from proMPO, and proteolytic processing of protomers and dimerization to yield mature MPO. Parallels between the biosynthesis of MPO and TPO as well as the impact of inherited mutations in the MPO gene on normal biosynthesis will be summarized. Lastly, specific gaps in our knowledge revealed by this review of our current understanding will be highlighted.
Collapse
|
36
|
Palladino END, Hartman CL, Albert CJ, Ford DA. The chlorinated lipidome originating from myeloperoxidase-derived HOCl targeting plasmalogens: Metabolism, clearance, and biological properties. Arch Biochem Biophys 2018; 641:31-38. [PMID: 29378164 DOI: 10.1016/j.abb.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 12/17/2022]
Abstract
Myeloperoxidase produces the two-electron oxidant HOCl, which targets plasmalogen phospholipids liberating 2-chlorofatty aldehyde. 2-Chlorofatty aldehyde has four known fates: 1) oxidation to 2-chlorofatty acid; 2) reduction to 2-chlorofatty alcohol; 3) Schiff base adduct formation with proteins and amines; and 4) reactivity with glutathione through nucleophilic attack of the α-chlorinated carbon. 2-Chlorofatty acid does not undergo conventional fatty acid β-oxidation due to the presence of the α-chlorinated carbon; however, 2-chlorofatty acid does undergo sequential ω-oxidation and β-oxidation from the ω-end, ultimately resulting in 2-chloroadipic acid urinary excretion. Recent studies have demonstrated that 2-chlorofatty acid clearance is increased by treatment with the PPAR-α agonist WY14643, which increases the enzymatic machinery responsible for hepatic ω-oxidation. Furthermore, 2-chlorofatty acid has been shown to be a PPAR-α agonist, and thus accelerates its own clearance. The roles of 2-chlorofatty aldehyde and 2-chlorofatty acid on leukocyte and endothelial function have been explored by several groups, suggesting that chlorinated lipids induce endothelial cell dysfunction, neutrophil chemotaxis, monocyte apoptosis, and alterations in vascular tone. Thus, the chlorinated lipidome, produced in response to leukocyte activation, is a potential biomarker and therapeutic target to modulate host response in inflammatory diseases.
Collapse
Affiliation(s)
- Elisa N D Palladino
- Edward A. Doisy Department of Biochemistry and Molecular Biology and the Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Celine L Hartman
- Edward A. Doisy Department of Biochemistry and Molecular Biology and the Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Carolyn J Albert
- Edward A. Doisy Department of Biochemistry and Molecular Biology and the Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology and the Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
37
|
Bernhart E, Kogelnik N, Prasch J, Gottschalk B, Goeritzer M, Depaoli MR, Reicher H, Nusshold C, Plastira I, Hammer A, Fauler G, Malli R, Graier WF, Malle E, Sattler W. 2-Chlorohexadecanoic acid induces ER stress and mitochondrial dysfunction in brain microvascular endothelial cells. Redox Biol 2018; 15:441-451. [PMID: 29413957 PMCID: PMC5975063 DOI: 10.1016/j.redox.2018.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 02/06/2023] Open
Abstract
Peripheral leukocytes induce blood-brain barrier (BBB) dysfunction through the release of cytotoxic mediators. These include hypochlorous acid (HOCl) that is formed via the myeloperoxidase-H2O2-chloride system of activated phagocytes. HOCl targets the endogenous pool of ether phospholipids (plasmalogens) generating chlorinated inflammatory mediators like e.g. 2-chlorohexadecanal and its conversion product 2-chlorohexadecanoic acid (2-ClHA). In the cerebrovasculature these compounds inflict damage to brain microvascular endothelial cells (BMVEC) that form the morphological basis of the BBB. To follow subcellular trafficking of 2-ClHA we synthesized a ‘clickable’ alkyne derivative (2-ClHyA) that phenocopied the biological activity of the parent compound. Confocal and superresolution structured illumination microscopy revealed accumulation of 2-ClHyA in the endoplasmic reticulum (ER) and mitochondria of human BMVEC (hCMEC/D3 cell line). 2-ClHA and its alkyne analogue interfered with protein palmitoylation, induced ER-stress markers, reduced the ER ATP content, and activated transcription and secretion of interleukin (IL)−6 as well as IL-8. 2-ClHA disrupted the mitochondrial membrane potential and induced procaspase-3 and PARP cleavage. The protein kinase R-like ER kinase (PERK) inhibitor GSK2606414 suppressed 2-ClHA-mediated activating transcription factor 4 synthesis and IL-6/8 secretion, but showed no effect on endothelial barrier dysfunction and cleavage of procaspase-3. Our data indicate that 2-ClHA induces potent lipotoxic responses in brain endothelial cells and could have implications in inflammation-induced BBB dysfunction.
Collapse
Affiliation(s)
- Eva Bernhart
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Nora Kogelnik
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Jürgen Prasch
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Madeleine Goeritzer
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Maria Rosa Depaoli
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Helga Reicher
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Christoph Nusshold
- Institute of Physiological Chemistry, Medical University of Graz, Austria.
| | - Ioanna Plastira
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Astrid Hammer
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Cell Biology, Histology and Embryology, Medical University of Graz, Austria.
| | - Günter Fauler
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria.
| | - Roland Malli
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| | - Ernst Malle
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Wolfgang Sattler
- Gottfried Schatz Research Center for Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| |
Collapse
|