1
|
Zhang S, Huang Y, Han C, Chen M, Yang Z, Wang C. Circulating mitochondria carrying cGAS promote endothelial Secreted group IIA phospholipase A2-mediated neuroinflammation through activating astroglial/microglial Integrin-alphavbeta3 in subfornical organ to augment central sympathetic overdrive in heart failure rats. Int Immunopharmacol 2025; 144:113649. [PMID: 39586230 DOI: 10.1016/j.intimp.2024.113649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Sympathoexcitation, a manifestation of heart-brain axis dysregulation, contributes to the progression of heart failure (HF). Our recent study revealed that circulating mitochondria (C-Mito), a newly identified mediator of multi-organ communication, promote sympathoexcitation in HF by aggravating endothelial cell (EC)-derived neuroinflammation in the subfornical organ (SFO), the cardiovascular autonomic neural center. The precise molecular mechanism by which C-Mito promotes SFO-induced endothelial neuroinflammation has not been fully elucidated. OBJECTIVE C-Mito carrying cGAS promote sympathoexcitation by targeting PLA2G2A in ECs of the SFO in HF rats. METHODS Male Sprague-Dawley (SD) rats received a subcutaneous injection of isoprenaline (ISO) at a dosage of 5 mg/kg/day for seven consecutive days to establish a HF model. C-Mito were isolated from HF rats and evaluated. The level of cGAS, a dsDNA sensor recently discovered to be directly localized on the outer membrane of mitochondria, was detected in C-Mito. C-Mito from HF rats (C-MitoHF) or control rats (C-MitoCtrl) were intravenously infused into HF rats. The accumulation of C-Mito in the ECs in the SFO was detected via double immunofluorescence staining. The SFO was processed for RNA sequencing (RNA-Seq) analysis. Secreted group IIA phospholipase A2 (PLA2G2A), the key gene involved in C-MitoHF-associated SFO dysfunction, was identified via bioinformatics analysis. Upregulation of PLA2G2A in the SFO ECs was assessed via immunofluorescence staining and immunoblotting, and PLA2G2A activity was evaluated. The interaction between cGAS and PLA2G2A was detected via co-immunoprecipitation. The dowstream molecular mechanisms of which PLA2G2A induced astroglial/microglial activation were also investigated. AAV9-TIE-shRNA (PLA2G2A) was introduced into the SFO to specifically knockdown endothelial PLA2G2A. Neuronal activation and glial proinflammatory polarization in the SFO were also evaluated. Renal sympathetic nerve activity (RSNA) was measured to evaluate central sympathetic output. Cardiac sympathetic hyperinnervation, myocardial remodeling, and left ventricular systolic function were assessed in C-Mito-treated HF rats. RESULTS Respiratory functional incompetence and oxidative damage were observed in C-MitoHF compared with C-MitoCtrl. Surprisingly, cGAS protein levels in C-MitoHF were significantly higher than those in C-MitoCtrl, while blocking cGAS with its specific inhibitor, RU.521, mitigated respiratory dysfunction and oxidative injury in C-MitoHF. C-Mito entered the ECs of the SFO in HF rats. RNA sequencing revealed that PLA2G2A is a key molecule for the induction of SFO dysfunction by C-MitoHF. The immunoblotting and immunofluorescence results confirmed that, compared with C-MitoCtrl, C-MitoHF increased endothelial PLA2G2A expression in the SFO of HF rats, which could be alleviated by attenuating C-MitoHF-localized cGAS. Furthermore, we found that cGAS directly interacts with PLA2G2A, increased the activity of PLA2AG2, which produced arachidonic acid, and also promoted PLA2G2A secretion in brain ECs. In addition, the inhibition of PLA2G2A in brain ECs significantly mitigated the proinflammatory effect of conditioned cell culture medium from C-MitoHF-treated ECs on astroglia and microglia. Also, we found that PLA2G2A secreted from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Specific knockdown of endothelial PLA2G2A in the SFO mitigated C-MitoHF-induced presympathetic neuronal sensitization, cardiac sympathetic hyperinnervation, RSNA activation, myocardial remodeling, and systolic dysfunction in HF rats. CONCLUSION C-Mito carrying cGAS promoted cardiac sympathoexcitation by directly targeting PLA2G2A in the ECs of the SFO in HF rats. Secreted PLA2G2A derived from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Our study indicated that inhibiting cGAS in C-Mito might be a potential treatment for central sympathetic overdrive in HF.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Yijun Huang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chengzhi Han
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Maoxiang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
2
|
Rahman MS, Hossain MS. Eicosanoids Signals in SARS-CoV-2 Infection: A Foe or Friend. Mol Biotechnol 2024; 66:3025-3041. [PMID: 37878227 DOI: 10.1007/s12033-023-00919-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
SARS-CoV-2 mediated infection instigated a scary pandemic state since 2019. They created havoc comprising death, imbalanced social structures, and a wrecked global economy. During infection, the inflammation and associated cytokine storm generate a critical pathological situation in the human body, especially in the lungs. By the passage of time of infection, inflammatory disorders, and multiple organ damage happen which might lead to death, if not treated properly. Until now, many pathological parameters have been used to understand the progress of the severity of COVID-19 but with limited success. Bioactive lipid mediators have the potential of initiating and resolving inflammation in any disease. The connection between lipid storm and inflammatory states of SARS-CoV-2 infection has surfaced and got importance to understand and mitigate the pathological states of COVID-19. As the role of eicosanoids in COVID-19 infection is not well defined, available information regarding this issue has been accumulated to address the possible network of eicosanoids related to the initiation of inflammation, promotion of cytokine storm, and resolution of inflammation, and highlight possible strategies for treatment and drug discovery related to SARS-CoV-2 infection in this study. Understanding the involvement of eicosanoids in exploration of cellular events provoked by SARS-CoV-2 infection has been summarized as an important factor to deescalate any upcoming catastrophe imposed by the lethal variants of this micro-monster. Additionally, this study also recognized the eicosanoid based drug discovery, treatment, and strategies for managing the severity of SARS-COV-2 infection.
Collapse
Affiliation(s)
- Mohammad Sharifur Rahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Mohammad Salim Hossain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh.
| |
Collapse
|
3
|
Li S, Zhang G, Hu J, Tian Y, Fu X. Ferroptosis at the nexus of metabolism and metabolic diseases. Theranostics 2024; 14:5826-5852. [PMID: 39346540 PMCID: PMC11426249 DOI: 10.7150/thno.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, is emerging as a crucial regulator of human physiology and pathology. Increasing evidence showcases a reciprocal relationship between ferroptosis and dysregulated metabolism, propagating a pathogenic vicious cycle that exacerbates pathology and human diseases, particularly metabolic disorders. Consequently, there is a rapidly growing interest in developing ferroptosis-based therapeutics. Therefore, a comprehensive understanding of the intricate interplay between ferroptosis and metabolism could provide an invaluable resource for mechanistic insight and therapeutic development. In this review, we summarize the important metabolic substances and associated pathways in ferroptosis initiation and progression, outline the cascade responses of ferroptosis in disease development, overview the roles and mechanisms of ferroptosis in metabolic diseases, introduce the methods for ferroptosis detection, and discuss the therapeutic perspectives of ferroptosis, which collectively aim to illustrate a comprehensive view of ferroptosis in basic, translational, and clinical science.
Collapse
Affiliation(s)
- Shuangwen Li
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guixiang Zhang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiankun Hu
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
4
|
Saballs M, Parra S, Martínez N, Amigo N, Cabau L, Iftimie S, Pavon R, Gabaldó X, Correig X, Paredes S, Vallvé JM, Castro A. Lipidomic and metabolomic changes in community-acquired and COVID-19 pneumonia. J Lipid Res 2024; 65:100622. [PMID: 39154734 PMCID: PMC11422144 DOI: 10.1016/j.jlr.2024.100622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024] Open
Abstract
This prospective observational study compared the 1H NMR blood lipidomes and metabolomes of 71 patients with community-acquired pneumonia (CAP), 75 patients with COVID-19 pneumonia, and 75 healthy controls (matched by age and sex) to identify potential biomarkers and pathways associated with respiratory infections. Both pneumonia groups had comparable severity indices, including mortality, invasive mechanical ventilation, and intensive care unit admission rates. Patients with COVID-19 pneumonia exhibited more pronounced hypolipidemia, with significantly lower levels of total cholesterol and LDL-c compared to patients with CAP. Atherogenic lipoprotein subclasses (VLDL-cholesterol, IDL-cholesterol, IDL-triglyceride, and LDL-triglyceride/LDL-cholesterol) were significantly increased in severe cases of both pneumonia types, while lower HDL-c and small, dense HDL particles were associated with more severe illness. Both infected groups showed decreased esterified cholesterol and increased triglycerides, along with reduced phosphatidylcholine, lysophosphatidylcholine, PUFA, omega-3 fatty acids, and DHA. Additionally, infected patients had elevated levels of glucose, lactate, 3-hydroxybutyrate, and acetone, which are linked to inflammation, hypoxemia, and sepsis. Increased levels of branched-chain amino acids, alanine, glycine, and creatine, which are involved in energy metabolism and protein catabolism, were also observed. Neurotransmitter synthesis metabolites like histidine and glutamate were higher in infected patients, especially those with COVID-19. Notably, severe infections showed a significant decrease in glutamine, essential for lymphocyte and macrophage energy. The severity of COVID-19 pneumonia was also associated with elevated glycoprotein levels (glycoprotein A, glycoprotein B, and glycoprotein F), indicating an inflammatory state. These findings suggest that metabolomic and lipidomic changes in pneumonia are connected to bioenergetic pathways regulating the immune response.
Collapse
Affiliation(s)
- Mireia Saballs
- Internal Medicine Department, Quiron Salud Hospital, Barcelona, Spain
| | - Sandra Parra
- Research Group of Autoimmunity, Infection and Thrombosis (GRAIIT), Pere Virgili for Health Research Institute (IISPV), Rovira and Virgili University (URV), Reus, Spain; Internal Medicine Department, "Sant Joan" University Hospital, Reus, Spain.
| | - Neus Martínez
- Biosfer Teslab, Reus, Spain; Department of Basic Medical Sciences, Rovira and Virgili University (URV), Pere Virgili for Health Research Institute (IISPV), Tarragona, Spain
| | - Nuria Amigo
- Biosfer Teslab, Reus, Spain; Department of Basic Medical Sciences, Rovira and Virgili University (URV), Pere Virgili for Health Research Institute (IISPV), Tarragona, Spain; Centre for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, Madrid, Spain
| | - Lydia Cabau
- Biosfer Teslab, Reus, Spain; Department of Basic Medical Sciences, Rovira and Virgili University (URV), Pere Virgili for Health Research Institute (IISPV), Tarragona, Spain
| | - Simona Iftimie
- Research Group of Autoimmunity, Infection and Thrombosis (GRAIIT), Pere Virgili for Health Research Institute (IISPV), Rovira and Virgili University (URV), Reus, Spain; Internal Medicine Department, "Sant Joan" University Hospital, Reus, Spain
| | - Raul Pavon
- Research Group of Autoimmunity, Infection and Thrombosis (GRAIIT), Pere Virgili for Health Research Institute (IISPV), Rovira and Virgili University (URV), Reus, Spain; Internal Medicine Department, "Sant Joan" University Hospital, Reus, Spain
| | - Xavi Gabaldó
- Research Group of Autoimmunity, Infection and Thrombosis (GRAIIT), Pere Virgili for Health Research Institute (IISPV), Rovira and Virgili University (URV), Reus, Spain; Clinical laboratory Department, "Sant Joan" University Hospital, Reus, Spain
| | - Xavier Correig
- Centre for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, Madrid, Spain; Department of Electronic Engineering, Rovira and Virgili University (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Silvia Paredes
- Rheumatology department, "Sant Joan" University Hospital, Reus, Spain
| | - Josep Maria Vallvé
- Centre for Biomedical Research Network on Diabetes and Associated Metabolic Diseases (CIBERDEM), ISCIII, Madrid, Spain; Lipids and Arteriosclerosis Research Unit, Rovira and Virgili University (URV), Reus, Spain; Pere Virgili for Health Research Institute (IISPV), Tarragona, Spain
| | - Antoni Castro
- Research Group of Autoimmunity, Infection and Thrombosis (GRAIIT), Pere Virgili for Health Research Institute (IISPV), Rovira and Virgili University (URV), Reus, Spain; Internal Medicine Department, "Sant Joan" University Hospital, Reus, Spain
| |
Collapse
|
5
|
Tsao FHC, Li Z, Amessoudji AW, Jawdat D, Sadat M, Arabi Y, Meyer KC. The Role of Serum Albumin and Secretory Phospholipase A2 in Sepsis. Int J Mol Sci 2024; 25:9413. [PMID: 39273360 PMCID: PMC11395451 DOI: 10.3390/ijms25179413] [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: 06/29/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Sepsis is caused by a dysregulated host response to an infection that leads to cascading cell death and eventually organ failure. In this study, the role of inflammatory response serum secretory phospholipase A2 (sPLA2) and albumin in sepsis was investigated by determining the activities of the two proteins in serial serum samples collected on different days from patients with sepsis after enrollment in the permissive underfeeding versus standard enteral feeding protocols in an intensive care unit. Serum sPLA2 and albumin showed an inverse relationship with increasing sPLA2 activity and decreasing albumin membrane-binding activity in patients with evolving complications of sepsis. The activities of sPLA2 and albumin returned to normal values more rapidly in the permissive underfeeding group than in the standard enteral feeding group. The inverse sPLA2-albumin activity relationship suggests a complex interplay between these two proteins and a regulatory mechanism underlying cell membrane phospholipid homeostasis in sepsis. The decreased albumin-membrane binding activity in patients' serum was due to its fatty acid-binding sites occupied by pre-bound fatty acids that might alter albumin's structure, binding capacities, and essential functions. The sPLA2-albumin dual serum assays may be useful in determining whether nutritional intervention effectively supports the more rapid recovery of appropriate immune responses in critically ill patients with sepsis.
Collapse
Affiliation(s)
- Francis H. C. Tsao
- Departments of Medicine, Division of Pulmonary and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA (K.C.M.)
| | - Zhanhai Li
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53792, USA;
| | - Amy W. Amessoudji
- Departments of Medicine, Division of Pulmonary and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA (K.C.M.)
| | - Dunia Jawdat
- Saudi Stem Cells Donor Registry and Cord Blood Bank, King Abdullah International Medical Research Center, College of medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | - Musharaf Sadat
- Intensive Care Department, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia; (M.S.); (Y.A.)
| | - Yaseen Arabi
- Intensive Care Department, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia; (M.S.); (Y.A.)
| | - Keith C. Meyer
- Departments of Medicine, Division of Pulmonary and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA (K.C.M.)
| |
Collapse
|
6
|
Mathews IT, Saminathan P, Henglin M, Liu M, Nadig N, Fang C, Mercader K, Chee SJ, Campbell AM, Patel AA, Tiwari S, Watrous JD, Ramesh K, Dicker M, Dao K, Meyer MA, Jousilahti P, Havulinna AS, Niiranen T, Salomaa V, Joosten LA, Netea MG, Zheng P, Kronenberg M, Patel SP, Gutkind JS, Ottensmeier C, Long T, Kaech SM, Hedrick CC, Cheng S, Jain M, Sharma S. Linoleoyl-lysophosphatidylcholine suppresses immune-related adverse events due to immune checkpoint blockade. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24310974. [PMID: 39148854 PMCID: PMC11326322 DOI: 10.1101/2024.08.07.24310974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Immune related adverse events (irAEs) after immune checkpoint blockade (ICB) therapy occur in a significant proportion of cancer patients. To date, the circulating mediators of ICB-irAEs remain poorly understood. Using non-targeted mass spectrometry, here we identify the circulating bio-active lipid linoleoyl-lysophosphatidylcholine (LPC 18:2) as a modulator of ICB-irAEs. In three independent human studies of ICB treatment for solid tumor, loss of circulating LPC 18:2 preceded the development of severe irAEs across multiple organ systems. In both healthy humans and severe ICB-irAE patients, low LPC 18:2 was found to correlate with high blood neutrophilia. Reduced LPC 18:2 biosynthesis was confirmed in preclinical ICB-irAE models, and LPC 18:2 supplementation in vivo suppressed neutrophilia and tissue inflammation without impacting ICB anti-tumor response. Results indicate that circulating LPC 18:2 suppresses human ICB-irAEs, and LPC 18:2 supplementation may improve ICB outcomes by preventing severe inflammation while maintaining anti-tumor immunity.
Collapse
Affiliation(s)
- Ian T. Mathews
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Mir Henglin
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mingyue Liu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | | | - Camille Fang
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Kysha Mercader
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | - Serena J. Chee
- University of Southampton, Southampton, United Kingdom
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | | | | | - Saumya Tiwari
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Jeramie D. Watrous
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Karthik Ramesh
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Khoi Dao
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | | | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki S. Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Genomics and Immunometabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Pan Zheng
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Molecular Biology, University of California San Diego, La Jolla CA 92093
| | - Sandip Pravin Patel
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
| | - J. Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
- Department of Pharmacology, University of California San Diego, La Jolla CA 92093
| | - Christian Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | - Tao Long
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Susan M. Kaech
- Salk Institute for Biological Studies, La Jolla CA 92037
| | - Catherine C. Hedrick
- Immunology Center of Georgia and Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Susan Cheng
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mohit Jain
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Sonia Sharma
- La Jolla Institute for Immunology, La Jolla, CA 92037
| |
Collapse
|
7
|
Hara A, Lu E, Johnstone L, Wei M, Sun S, Hallmark B, Watkins JC, Zhang HH, Yao G, Chilton FH. Identification of an Allele-Specific Transcription Factor Binding Interaction that May Regulate PLA2G2A Gene Expression. Bioinform Biol Insights 2024; 18:11779322241261427. [PMID: 39081667 PMCID: PMC11287738 DOI: 10.1177/11779322241261427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/24/2024] [Indexed: 08/02/2024] Open
Abstract
The secreted phospholipase A2 (sPLA2) isoform, sPLA2-IIA, has been implicated in a variety of diseases and conditions, including bacteremia, cardiovascular disease, COVID-19, sepsis, adult respiratory distress syndrome, and certain cancers. Given its significant role in these conditions, understanding the regulatory mechanisms impacting its levels is crucial. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs), including rs11573156, that are associated with circulating levels of sPLA2-IIA. The work in the manuscript leveraged 4 publicly available datasets to investigate the mechanism by which rs11573156 influences sPLA2-IIA levels via bioinformatics and modeling analysis. Through genotype-tissue expression (GTEx), 234 expression quantitative trait loci (eQTLs) were identified for the gene that encodes for sPLA2-IIA, PLA2G2A. SNP2TFBS was used to ascertain the binding affinities between transcription factors (TFs) to both the reference and alternative alleles of identified eQTL SNPs. Subsequently, candidate TF-SNP interactions were cross-referenced with the ChIP-seq results in matched tissues from ENCODE. SP1-rs11573156 emerged as the significant TF-SNP pair in the liver. Further analysis revealed that the upregulation of PLA2G2A transcript levels through the rs11573156 variant was likely affected by tissue SP1 protein levels. Using an ordinary differential equation based on Michaelis-Menten kinetic assumptions, we modeled the dependence of PLA2G2A transcription on SP1 protein levels, incorporating the SNP influence. Collectively, our analysis strongly suggests that the difference in the binding dynamics of SP1 to different rs11573156 alleles may underlie the allele-specific PLA2G2A expression in different tissues, a mechanistic model that awaits future direct experimental validation. This mechanism likely contributes to the variation in circulating sPLA2-IIA protein levels in the human population, with implications for a wide range of human diseases.
Collapse
Affiliation(s)
- Aki Hara
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
| | - Eric Lu
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Laurel Johnstone
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
| | - Michelle Wei
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Shudong Sun
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Brian Hallmark
- BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - Joseph C Watkins
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Hao Helen Zhang
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Guang Yao
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, USA
- Center for Precision Nutrition and Wellness, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
8
|
Pethe A, Joshi S, Ali Dar T, Poddar NK. Revisiting the role of phospholipases in alzheimer's: crosstalk with processed food. Crit Rev Food Sci Nutr 2024:1-19. [PMID: 39002140 DOI: 10.1080/10408398.2024.2377290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
Phospholipases such as phospholipase-A, phospholipase-B, phospholipase-C and phospholipase-D are important functional enzymes of the cell membrane responsible for a variety of functions such as signal transduction, production of lipid mediators, metabolite digestion and playing a pathological role in central nervous system diseases. Phospholipases have shown an association with Alzheimer's disease and these enzymes have found a correlation with several metabolic pathways that can lead to the activation of inflammatory signals via astrocytes and microglial cells. We also highlighted unhealthy practices like smoking and consuming processed foods, rich in nitroso compounds and phosphatidic acid, which contribute to neuronal damage in AD through phospholipases. A few therapeutic approaches such as the use of inhibitors of phospholipase-D,phospholipase A2 as well as autophagy-mediated inhibition have been discussed to control the onset of AD. This paper serves as a crosstalk between phospholipases and their role in neurodegenerative pathways as well as their influence on other biomolecules of lipid membranes, which are acquired through unhealthy diets and possible methods to treat these anomalies occurring due to their metabolic disorder involving phospholipases acting as major signaling molecules.
Collapse
Affiliation(s)
- Atharv Pethe
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Siddhi Joshi
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Nitesh Kumar Poddar
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| |
Collapse
|
9
|
Mönki J, Mykkänen A. Lipids in Equine Airway Inflammation: An Overview of Current Knowledge. Animals (Basel) 2024; 14:1812. [PMID: 38929431 PMCID: PMC11200544 DOI: 10.3390/ani14121812] [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: 05/22/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Mild-moderate and severe equine asthma (MEA and SEA) are prevalent inflammatory airway conditions affecting horses of numerous breeds and disciplines. Despite extensive research, detailed disease pathophysiology and the differences between MEA and SEA are still not completely understood. Bronchoalveolar lavage fluid cytology, broadly used in clinical practice and in equine asthma research, has limited means to represent the inflammatory status in the lower airways. Lipidomics is a field of science that can be utilized in investigating cellular mechanisms and cell-to-cell interactions. Studies in lipidomics have a broad variety of foci, of which fatty acid and lipid mediator profile analyses and global lipidomics have been implemented in veterinary medicine. As many crucial proinflammatory and proresolving mediators are lipids, lipidomic studies offer an interesting yet largely unexplored means to investigate inflammatory reactions in equine airways. The aim of this review article is to collect and summarize the findings of recent lipidomic studies on equine airway inflammation.
Collapse
Affiliation(s)
| | - Anna Mykkänen
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Viikintie 49, P.O. Box 57, 00014 Helsinki, Finland;
| |
Collapse
|
10
|
Bai X, Wang S, Shu L, Cao Q, Hu H, Zhu Y, Chen C. Hawthorn leaf flavonoids alleviate the deterioration of atherosclerosis by inhibiting SCAP-SREBP2-LDLR pathway through sPLA2-ⅡA signaling in macrophages in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118006. [PMID: 38442806 DOI: 10.1016/j.jep.2024.118006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hawthorn leaves are a combination of the dried leaves of the Rosaceae plants, i.e., Crataegus pinnatifida Bge. or Crataegus pinnatifida Bge. var. major N. E. Br., is primarily cultivated in East Asia, North America, and Europe. hawthorn leaf flavonoids (HLF) are the main part of extraction. The HLF have demonstrated potential in preventing hypertension, inflammation, hyperlipidemia, and atherosclerosis. However, the potential pharmacological mechanism behind its anti-atherosclerotic effect has yet to be explored. AIM OF THE STUDY The in vivo and in vitro effects of HLF on lipid-mediated foam cell formation were investigated, with a specific focus on the levels of secreted phospholipase A2 type IIA (sPLA2-II A) in macrophage cells. MATERIALS AND METHODS The primary constituents of HLF were analyzed using ultra-high performance liquid chromatography and liquid chromatography-tandem mass spectrometry. In vivo, HLF, at concentrations of 5 mg/kg, 20 mg/kg, and 40 mg/kg, were administered to apolipoprotein E knockout mice (ApoE-/-) fed by high-fat diet (HFD) for 16 weeks. Aorta and serum samples were collected to identify lesion areas and lipids through mass spectrometry analysis to dissect the pathological process. RAW264.7 cells were incubated with oxidized low-density lipoprotein (ox-LDL) alone, or ox-LDL combined with different doses of HLF (100, 50, and 25 μg/ml), or ox-LDL plus 24-h sPLA2-IIA inhibitors, for cell biology analysis. Lipids and inflammatory cytokines were detected using biochemical analyzers and ELISA, while plaque size and collagen content of plaque were assessed by HE and the Masson staining of the aorta. The lipid deposition in macrophages was observed by Oil Red O staining. The expression of sPLA2-IIA and SCAP-SREBP2-LDLR was determined by RT-qPCR and Western blot analysis. RESULTS The chemical profile of HLF was studied using UPLC-Q-TOF-MS/MS, allowing the tentative identification of 20 compounds, comprising 1 phenolic acid, 9 flavonols and 10 flavones, including isovitexin, vitexin-4″-O-glucoside, quercetin-3-O-robibioside, rutin, vitexin-2″-O-rhamnoside, quercetin, etc. HLF decreased total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C) levels in ApoE-/- mice (P < 0.05), reduced ox-LDL uptake, inhibited level of inflammatory factors, such as IL-6, IL-8, TNF-α, and IL-1ꞵ (P < 0.001), and alleviated aortic plaques with a thicker fibrous cap. HLF effectively attenuated foam cell formation in ox-LDL-treated RAW264.7 macrophages, and reduced levels of intracellular TC, free cholesterol (FC), cholesteryl ester (CE), IL-6, TNF-α, and IL-1β (P < 0.001). In both in vivo and in vitro experiments, HLF significantly downregulated the expression of sPLA2-IIA, SCAP, SREBP2, LDLR, HMGCR, and LOX-1 (P < 0.05). Furthermore, sPLA2-IIA inhibitor effectively mitigated inflammatory release in RAW264.7 macrophages and regulated SCAP-SREBP2-LDLR signaling pathway by inhibiting sPLA2-IIA secretion (P < 0.05). CONCLUSION HLF exerted a protective effect against atherosclerosis through inhibiting sPLA2-IIA to diminish SCAP-SREBP2-LDLR signaling pathway, to reduce LDL uptake caused foam cell formation, and to slow down the progression of atherosclerosis in mice.
Collapse
Affiliation(s)
- Xufeng Bai
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shuwen Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Limei Shu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qingyu Cao
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Huiming Hu
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China; Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi, 330052, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| | - Yanchen Zhu
- College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
11
|
Greiner JV, Glonek T. Phospholipid analyses of rabbit ocular surface tissues. Exp Eye Res 2024; 243:109911. [PMID: 38663719 DOI: 10.1016/j.exer.2024.109911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
The tissues of the integument covering the ocular surface comprise a mucus membrane functioning as a protective physical barrier and has the ability to mount a defensive inflammatory response. Since lipid metabolism has a role in both of these functions, we studied normal membrane phospholipids (PL) of the cornea and bulbar conjunctiva to (1) determine baseline PL profiles of these tissues, (2) compare and contrast these individual PL metabolite profiles as well as groups of metabolites, and (3) describe pathway-specific metabolic interrelations among these tissues. Corneal and conjunctival tissue samples were isolated from rabbit eyes (n = 30) and extracted with chloroform-methanol using a modified Folch procedure. 31P nuclear magnetic resonance spectroscopy was used to qualitatively and quantitatively measure tissue PL profiles. The cornea and conjunctiva, respectively, have the following PL composition (mole % of total detected phospholipid): phosphatidylglycerol (PG) -, 0.4; lysophosphatidylethanolamine 1.2, -; phosphatidic acid -, 0.4; diPG (cardiolipin) 2.1, 3.5; unknown PL at the chemical shift of 0.13 δ 1.5, 0.9; ethanolamine plasmalogen 11.2, 13.0; phosphatidylethanolamine 11.5, 12.8; phosphatidylserine 8.9, 10.1; sphingomyelin 10.2, 10.7; lysophosphatidylcholine 0.9, 1.4; phosphatidylinositol 5.3, 5.3; phosphatidylcholine (PC) plasmalogen or alkylacylPC 2.2, 1.9; PC 45.1, 40.0. In addition, 28 PL metabolic indices were calculated from these data, which permitted pathway-specific lipid analyses. This study (1) establishes PL profiles of the two ocular tissues of the integument that cover the surface of the eye, (2) compares and contrasts indices comprised of ratios and combinations of PL, and (3) describes pathway-specific metabolic interrelations among these tissues to serve as baselines for studies involving the distribution of tissue phospholipids.
Collapse
Affiliation(s)
- Jack V Greiner
- Department of Ophthalmology, Harvard Medical School, 243 Charles St., Boston, MA, USA; Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, 20 Staniford St., Boston, MA, USA; Clinical Eye Research of Boston, 5 Whittier Pl, Ste. 102, Boston, MA, USA; Magnetic Resonance Laboratory, Chicago College of Osteopathic Medicine, Chicago, 5200 S. Ellis Ave., IL, USA.
| | - Thomas Glonek
- Clinical Eye Research of Boston, 5 Whittier Pl, Ste. 102, Boston, MA, USA; Magnetic Resonance Laboratory, Chicago College of Osteopathic Medicine, Chicago, 5200 S. Ellis Ave., IL, USA.
| |
Collapse
|
12
|
Bu H, Zhang S, Li P, Liu Z, Liu Y, Li Z, Liu X, Wang Z, Feng L, Chen L, Qu L. Secreted phospholipase PLA2G2E contributes to regulation of T cell immune response against influenza virus infection. J Virol 2024; 98:e0019824. [PMID: 38591879 PMCID: PMC11092358 DOI: 10.1128/jvi.00198-24] [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: 02/07/2024] [Accepted: 03/24/2024] [Indexed: 04/10/2024] Open
Abstract
The involvement of secreted phospholipase A2s in respiratory diseases, such as asthma and respiratory viral infections, is well-established. However, the specific role of secreted phospholipase A2 group IIE (PLA2G2E) during influenza virus infection remains unexplored. Here, we investigated the role of PLA2G2E during H1N1 influenza virus infection using a targeted mouse model lacking Pla2g2e gene (Pla2g2e-/-). Our findings demonstrated that Pla2g2e-/- mice had significantly lower survival rates and higher viral loads in lungs compared to wild-type mice following influenza virus infection. While Pla2g2e-/- mice displayed comparable innate and humoral immune responses to influenza virus challenge, the animals showed impaired influenza-specific cellular immunity and reduced T cell-mediated cytotoxicity. This indicates that PLA2G2E is involved in regulating specific T cell responses during influenza virus infection. Furthermore, transgenic mice expressing the human PLA2G2E gene exhibited resistance to influenza virus infection along with enhanced influenza-specific cellular immunity and T cell-mediated cytotoxicity. Pla2g2e deficiency resulted in perturbation of lipid mediators in the lung and T cells, potentially contributing to its impact on the anti-influenza immune response. Taken together, these findings suggest that targeting PLA2G2E could hold potential as a therapeutic strategy for managing influenza virus infections.IMPORTANCEThe influenza virus is a highly transmissible respiratory pathogen that continues to pose a significant public health concern. It effectively evades humoral immune protection conferred by vaccines and natural infection due to its continuous viral evolution through the genetic processes of antigenic drift and shift. Recognition of conserved non-mutable viral epitopes by T cells may provide broad immunity against influenza virus. In this study, we have demonstrated that phospholipase A2 group IIE (PLA2G2E) plays a crucial role in protecting against influenza virus infection through the regulation of T cell responses, while not affecting innate and humoral immune responses. Targeting PLA2G2E could therefore represent a potential therapeutic strategy for managing influenza virus infection.
Collapse
MESH Headings
- Animals
- Mice
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Influenza A Virus, H1N1 Subtype/immunology
- Lung/virology
- Lung/immunology
- Lung/pathology
- Humans
- Group II Phospholipases A2/genetics
- Group II Phospholipases A2/immunology
- T-Lymphocytes/immunology
- Mice, Knockout
- Immunity, Cellular
- Mice, Inbred C57BL
- Mice, Transgenic
- Viral Load
- Disease Models, Animal
- Immunity, Humoral
- Immunity, Innate
- Influenza, Human/immunology
- Influenza, Human/virology
- Female
Collapse
Affiliation(s)
- Hemeng Bu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shengnan Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zijian Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yichu Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhixia Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xinglong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhi Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
13
|
Koganesawa M, Dwyer D, Alhallak K, Nagai J, Zaleski K, Samuchiwal S, Hiroaki H, Nishida A, Hirsch TI, Brennan PJ, Puder M, Balestrieri B. Pla2g5 contributes to viral-like-induced lung inflammation through macrophage proliferation and LA/Ffar1 lung cell recruitment. Immunology 2024; 172:144-162. [PMID: 38361249 PMCID: PMC11057362 DOI: 10.1111/imm.13766] [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: 09/26/2023] [Accepted: 02/01/2024] [Indexed: 02/17/2024] Open
Abstract
Macrophages expressing group V phospholipase A2 (Pla2g5) release the free fatty acid (FFA) linoleic acid (LA), potentiating lung type 2 inflammation. Although Pla2g5 and LA increase in viral infections, their role remains obscure. We generated Pla2g5flox/flox mice, deleted Pla2g5 by using the Cx3cr1cre transgene, and activated bone marrow-derived macrophages (BM-Macs) with poly:IC, a synthetic double-stranded RNA that triggers a viral-like immune response, known Pla2g5-dependent stimuli (IL-4, LPS + IFNγ, IL-33 + IL-4 + GM-CSF) and poly:IC + LA followed by lipidomic and transcriptomic analysis. Poly:IC-activated Pla2g5flox/flox;Cx3cr1cre/+ BM-Macs had downregulation of major bioactive lipids and critical enzymes producing those bioactive lipids. In addition, AKT phosphorylation was lower in poly:IC-stimulated Pla2g5flox/flox;Cx3cr1cre/+ BM-Macs, which was not restored by adding LA to poly:IC-stimulated BM-Macs. Consistently, Pla2g5flox/flox;Cx3cr1cre/+ mice had diminished poly:IC-induced lung inflammation, including inflammatory macrophage proliferation, while challenging Pla2g5flox/flox;Cx3cr1cre/+ mice with poly:IC + LA partially restored lung inflammation and inflammatory macrophage proliferation. Finally, mice lacking FFA receptor-1 (Ffar1)-null mice had reduced poly:IC-induced lung cell recruitment and tissue macrophage proliferation, not corrected by LA. Thus, Pla2g5 contributes to poly:IC-induced lung inflammation by regulating inflammatory macrophage proliferation and LA/Ffar1-mediated lung cell recruitment and tissue macrophage proliferation.
Collapse
Affiliation(s)
- Masaya Koganesawa
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Daniel Dwyer
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Kinan Alhallak
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Jun Nagai
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Kendall Zaleski
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Sachin Samuchiwal
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Hayashi Hiroaki
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Airi Nishida
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Thomas I. Hirsch
- Department of Surgery and Vascular Biology Program Boston Children’s Hospital, Boston, MA
| | - Patrick J. Brennan
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| | - Mark Puder
- Department of Surgery and Vascular Biology Program Boston Children’s Hospital, Boston, MA
| | - Barbara Balestrieri
- Division of Allergy and Clinical Immunology, Vinik Center for Translational Immunology Research, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
14
|
Malavige GN, Ogg GS. Molecular mechanisms in the pathogenesis of dengue infections. Trends Mol Med 2024; 30:484-498. [PMID: 38582622 DOI: 10.1016/j.molmed.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Dengue is the most rapidly emerging climate-sensitive infection, and morbidity/mortality and disease incidence are rising markedly, leading to healthcare systems being overwhelmed. There are currently no specific treatments for dengue or prognostic markers to identify those who will progress to severe disease. Owing to an increase in the burden of illness and a change in epidemiology, many patients experience severe disease. Our limited understanding of the complex mechanisms of disease pathogenesis has significantly hampered the development of safe and effective treatments, vaccines, and biomarkers. We discuss the molecular mechanisms of dengue pathogenesis, the gaps in our knowledge, and recent advances, as well as the most crucial questions to be answered to enable the development of therapeutics, biomarkers, and vaccines.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka; Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| | - Graham S Ogg
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka; Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Ji X, Ji HL. Metabolic signatures of acute respiratory distress syndrome: COVID versus non-COVID. Am J Physiol Lung Cell Mol Physiol 2024; 326:L596-L603. [PMID: 38469648 PMCID: PMC11380973 DOI: 10.1152/ajplung.00266.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a fatal pulmonary disorder characterized by severe hypoxia and inflammation. ARDS is commonly triggered by systemic and pulmonary infections, with bacteria and viruses. Notable pathogens include Pseudomonas aeruginosa, Streptococcus aureus, Enterobacter species, coronaviruses, influenza viruses, and herpesviruses. COVID-19 ARDS represents the latest etiological phenotype of the disease. The pathogenesis of ARDS caused by bacteria and viruses exhibits variations in host immune responses and lung mesenchymal injury. We postulate that the systemic and pulmonary metabolomics profiles of ARDS induced by COVID-19 pathogens may exhibit distinctions compared with those induced by other infectious agents. This review aims to compare metabolic signatures in blood and lung specimens specifically within the context of ARDS. Both prevalent and phenotype-specific metabolomic signatures, including but not limited to glycolysis, ketone body production, lipid oxidation, and dysregulation of the kynurenine pathways, were thoroughly examined in this review. The distinctions in metabolic signatures between COVID-19 and non-COVID ARDS have the potential to reveal new biomarkers, elucidate pathogenic mechanisms, identify druggable targets, and facilitate differential diagnosis in the future.
Collapse
Affiliation(s)
- Xiangming Ji
- Department of Nutrition, Georgia State University, Atlanta, Georgia, United States
| | - Hong-Long Ji
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, Illinois, United States
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, Illinois, United States
| |
Collapse
|
16
|
Lu E, Hara A, Sun S, Hallmark B, Snider JM, Seeds MC, Watkins JC, McCall CE, Zhang HH, Yao G, Chilton FH. Temporal associations of plasma levels of the secreted phospholipase A 2 family and mortality in severe COVID-19. Eur J Immunol 2024:e2350721. [PMID: 38651231 DOI: 10.1002/eji.202350721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 04/25/2024]
Abstract
Previous research suggests that group IIA-secreted phospholipase A2 (sPLA2-IIA) plays a role in and predicts lethal COVID-19 disease. The current study reanalyzed a longitudinal proteomic data set to determine the temporal relationship between levels of several members of a family of sPLA2 isoforms and the severity of COVID-19 in 214 ICU patients. The levels of six secreted PLA2 isoforms, sPLA2-IIA, sPLA2-V, sPLA2-X, sPLA2-IB, sPLA2-IIC, and sPLA2-XVI, increased over the first 7 ICU days in those who succumbed to the disease but attenuated over the same time period in survivors. In contrast, a reversed pattern in sPLA2-IID and sPLA2-XIIB levels over 7 days suggests a protective role of these two isoforms. Furthermore, decision tree models demonstrated that sPLA2-IIA outperformed top-ranked cytokines and chemokines as a predictor of patient outcome. Taken together, proteomic analysis revealed temporal sPLA2 patterns that reflect the critical roles of sPLA2 isoforms in severe COVID-19 disease.
Collapse
Affiliation(s)
- Eric Lu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| | - Aki Hara
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, USA
| | - Shudong Sun
- Department of Mathematics, University of Arizona, Tucson, Arizona, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
| | - Brian Hallmark
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Justin M Snider
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, USA
- Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
- Center for Precision Nutrition and Wellness, University of Arizona, Tucson, Arizona, USA
| | - Michael C Seeds
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Joseph C Watkins
- Department of Mathematics, University of Arizona, Tucson, Arizona, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
| | - Charles E McCall
- Departments of Internal Medicine, Microbiology and Immunology, and Clinical and Translational Sciences Institute, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Hao Helen Zhang
- Department of Mathematics, University of Arizona, Tucson, Arizona, USA
- Statistics Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
| | - Guang Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
- Center for Precision Nutrition and Wellness, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
17
|
Radwan E, Abdelaziz A, Mandour MAM, Meki ARMA, El-Kholy MM, Mohamed MN. MBOAT7 expression is associated with disease progression in COVID-19 patients. Mol Biol Rep 2024; 51:79. [PMID: 38183501 PMCID: PMC10771377 DOI: 10.1007/s11033-023-09009-9] [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: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND AND AIM The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in late 2019 caused a pandemic of acute respiratory disease, named coronavirus disease 2019 (COVID-19). COVID-19 became one of the most challenging health emergencies, hence the necessity to find different prognostic factors for disease progression, and severity. Membrane bound O-acyltransferase domain containing 7 (MBOAT7) demonstrates anti-inflammatory effects through acting as a fine-tune regulator of the amount of cellular free arachidonic acid. We aimed in this study to evaluate MBOAT7 expression in COVID-19 patients and to correlate it with disease severity and outcomes. METHODS This case-control study included 56 patients with confirmed SARS-CoV-2 diagnosis and 28 control subjects. Patients were further classified into moderate (n = 28) and severe (n = 28) cases. MBOAT7, tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß) mRNA levels were evaluated in peripheral blood mononuclear cells (PBMC) samples isolated from patients and control subjects by real time quantitative polymerase chain reaction (RT-qPCR). In addition, circulating MBOAT7 protein levels were assayed by enzyme-linked immunosorbent assay (ELISA). RESULTS Significant lower levels of circulating MBOAT7 mRNA and protein were observed in COVID-19 patients compared to control subjects with severe COVID-19 cases showing significant lower levels compared to moderate cases. Moreover, severe cases showed a significant upregulation of TNF-α and IL-1ß mRNA. MBOAT7 mRNA and protein levels were significantly correlated with inflammatory markers (TNF-α, IL-1ß, C-reactive protein (CRP), and ferritin), liver enzymes, severity, and oxygen saturation levels. CONCLUSION COVID-19 is associated with downregulation of MBAOT7, which correlates with disease severity.
Collapse
Affiliation(s)
- Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt.
| | - Ahmed Abdelaziz
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt
| | - Manal A M Mandour
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Abdel-Raheim M A Meki
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
- Department of Biochemistry, Sphinx University, New Assiut City, Assiut 10, Egypt
| | - Maha M El-Kholy
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwan N Mohamed
- Department of Chest diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
18
|
Wang X, Ding D, Liu Y, Zhao H, Sun J, Li Y, Cao J, Hou S, Zhang Y. Plasma lipidome reveals susceptibility and resistance of Pekin ducks to DHAV-3. Int J Biol Macromol 2023; 253:127095. [PMID: 37758112 DOI: 10.1016/j.ijbiomac.2023.127095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 09/23/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Duck hepatitis A virus genotype 3 (DHAV-3) is the most popular pathogen of duck viral hepatitis (DVH) and has led to a huge economic threat to the Asian duck industry. In this work, we investigated the differences in the LC-MS/MS-based dynamic lipid profiles between susceptible and resistant Pekin duck lines with DHAV-3 infection. We found that the plasma lipidome of the two duck lines was characterized differently in expression levels of lipids during the infection, such as decreased levels of glycerolipids and increased levels of cholesteryl esters and glycerophospholipids in susceptible ducks compared with resistant ducks. By integrating lipidomics and transcriptomics analysis, we showed that the altered homeostasis of lipids was potentially regulated by a variety of differentially expressed genes including CHPT1, PI4K2A, and OSBP2 between the two duck lines, which could account for liver dysfunction, apoptosis, and illness upon DHAV-3 infection. Using the least absolute shrinkage and selection operator (LASSO) approach, we determined a total of 25 infection-related lipids that were able to distinguish between the infection states of susceptible and resistant ducks. This study provides molecular clues for elucidating the pathogenesis and therapeutic strategies of DHAV-3 infection in ducklings, which has implication for the development of resistance breeding.
Collapse
Affiliation(s)
- Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Dingbang Ding
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haonan Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jianfeng Sun
- Botnar Research Centre, University of Oxford, OX3 7LD Oxford, United Kingdom
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junting Cao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuisheng Hou
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yunsheng Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
19
|
Hara A, Lu E, Johnstone L, Wei M, Sun S, Hallmark B, Watkins JC, Zhang HH, Yao G, Chilton FH. Identification of an allele-specific transcription factor binding interaction that regulates PLA2G2A gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571290. [PMID: 38168258 PMCID: PMC10760018 DOI: 10.1101/2023.12.12.571290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The secreted phospholipase A 2 (sPLA 2 ) isoform, sPLA 2 -IIA, has been implicated in a variety of diseases and conditions, including bacteremia, cardiovascular disease, COVID-19, sepsis, adult respiratory distress syndrome, and certain cancers. Given its significant role in these conditions, understanding the regulatory mechanisms impacting its levels is crucial. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs), including rs11573156, that are associated with circulating levels of sPLA 2 -IIA. Through Genotype-Tissue Expression (GTEx), 234 expression quantitative trait loci (eQTLs) were identified for the gene that encodes for sPLA 2 -IIA, PLA2G2A . SNP2TFBS ( https://ccg.epfl.ch/snp2tfbs/ ) was utilized to ascertain the binding affinities between transcription factors (TFs) to both the reference and alternative alleles of identified SNPs. Subsequently, ChIP-seq peaks highlighted the TF combinations that specifically bind to the SNP, rs11573156. SP1 emerged as a significant TF/SNP pair in liver cells, with rs11573156/SP1 interaction being most prominent in liver, prostate, ovary, and adipose tissues. Further analysis revealed that the upregulation of PLA2G2A transcript levels through the rs11573156 variant was affected by tissue SP1 protein levels. By leveraging an ordinary differential equation, structured upon Michaelis-Menten enzyme kinetics assumptions, we modeled the PLA2G2A transcription's dependence on SP1 protein levels, incorporating the SNP's influence. Collectively, these data strongly suggest that the binding affinity differences of SP1 for the different rs11573156 alleles can influence PLA2G2A expression. This, in turn, can modulate sPLA2-IIA levels, impacting a wide range of human diseases.
Collapse
|
20
|
Potashnikova DM, Tvorogova AV, Saidova AA, Sotnikova TN, Arifulin EA, Lipina TV, Shirokova OM, Melnikov ES, Rodina TA, Valyaeva AA, Zharikova AA, Zayratyants GO, Zayratyants OV, Sheval EV, Vasilieva EJ. Lung inflammation is associated with lipid deposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2022.12.30.522299. [PMID: 36789445 PMCID: PMC9928036 DOI: 10.1101/2022.12.30.522299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lung inflammation, pneumonia, is an acute respiratory disease of varying etiology that has recently drawn much attention during the COVID-19 pandemic as lungs are among the main targets for SARS-CoV-2. Multiple other etiological agents are associated with pneumonias. Here, we describe a newly-recognized pathology, namely abnormal lipid depositions in the lungs of patients who died from COVID-19 as well as from non-COVID-19 pneumonias. Our analysis of both semi-thin and Sudan III-stained lung specimens revealed extracellular and intracellular lipid depositions irrespective of the pneumonia etiology. Most notably, lipid depositions were located within vessels adjacent to inflamed regions, where they apparently interfere with the blood flow. Structurally, the lipid droplets in the inflamed lung tissue were homogeneous and lacked outer membranes as assessed by electron microscopy. Morphometric analysis of lipid droplet deposition area allowed us to distinguish the non-pneumonia control lung specimens from the macroscopically intact area of the pneumonia lung and from the inflamed area of the pneumonia lung. Our measurements revealed a gradient of lipid deposition towards the inflamed region. The pattern of lipid distribution proved universal for all pneumonias. Finally, lipid metabolism in the lung tissue was assessed by the fatty acid analysis and by expression of genes involved in lipid turnover. Chromato-mass spectrometry revealed that unsaturated fatty acid content was elevated at inflammation sites compared to that in control non-inflamed lung tissue from the same individual. The expression of genes involved in lipid metabolism was altered in pneumonia, as shown by qPCR and in silico RNA-seq analysis. Thus, pneumonias of various etiologies are associated with specific lipid abnormalities; therefore, lipid metabolism can be considered to be a target for new therapeutic strategies.
Collapse
|
21
|
Mazurak VC, Rivas-Serna IM, Parsons SR, Monirujjaman M, Maybank KE, Woo SK, Rewa OG, Cave AJ, Richard C, Clandinin MT. Plasma essential fatty acid on hospital admission is a marker of COVID-19 disease severity. Sci Rep 2023; 13:18973. [PMID: 37923927 PMCID: PMC10624896 DOI: 10.1038/s41598-023-46247-0] [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: 06/15/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
It is important for allocation of resources to predict those COVID patients at high risk of dying or organ failure. Early signals to initiate cellular events of host immunity can be derived from essential fatty acid metabolites preceding the cascade of proinflammatory signals. Much research has focused on understanding later proinflammatory responses. We assessed if remodelling of plasma phospholipid content of essential fatty acids by the COVID-19 virus provides early markers for potential death and disease severity. Here we show that, at hospital admission, COVID-19 infected subjects who survive exhibit higher proportions of C20:4n-6 in plasma phospholipids concurrent with marked proinflammatory cytokine elevation in plasma compared to healthy subjects. In contrast, more than half of subjects who die of this virus exhibit very low C18:2n-6 and C20:4n-6 content in plasma phospholipids on hospital admission compared with healthy control subjects. Moreover, in these subjects who die, the low level of primary inflammatory signals indicates limited or aberrant stimulation of host immunity. We conclude that COVID-19 infection results in early fundamental remodelling of essential fatty acid metabolism. In subjects with high mortality, it appears that plasma n-6 fatty acid content is too low to stimulate cellular events of host immunity.
Collapse
Affiliation(s)
- Vera C Mazurak
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Irma Magaly Rivas-Serna
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Sarah R Parsons
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Md Monirujjaman
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Krista E Maybank
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Stanley K Woo
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Oleksa G Rewa
- Department of Critical Care Medicine, Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Andrew J Cave
- Department of Family Medicine, University of Alberta, Edmonton, T6G 2P5, Canada
| | - Caroline Richard
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada
| | - M Thomas Clandinin
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2P5, Canada.
- Department of Medicine, University of Alberta, Edmonton, T6G 2P5, Canada.
| |
Collapse
|
22
|
Meng H, Sengupta A, Ricciotti E, Mrčela A, Mathew D, Mazaleuskaya LL, Ghosh S, Brooks TG, Turner AP, Schanoski AS, Lahens NF, Tan AW, Woolfork A, Grant G, Susztak K, Letizia AG, Sealfon SC, Wherry EJ, Laudanski K, Weljie AM, Meyer NJ, FitzGerald GA. Deep phenotyping of the lipidomic response in COVID-19 and non-COVID-19 sepsis. Clin Transl Med 2023; 13:e1440. [PMID: 37948331 PMCID: PMC10637636 DOI: 10.1002/ctm2.1440] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/15/2023] [Accepted: 10/01/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Lipids may influence cellular penetrance by viral pathogens and the immune response that they evoke. We deeply phenotyped the lipidomic response to SARs-CoV-2 and compared that with infection with other pathogens in patients admitted with acute respiratory distress syndrome to an intensive care unit (ICU). METHODS Mass spectrometry was used to characterise lipids and relate them to proteins, peripheral cell immunotypes and disease severity. RESULTS Circulating phospholipases (sPLA2, cPLA2 (PLA2G4A) and PLA2G2D) were elevated on admission in all ICU groups. Cyclooxygenase, lipoxygenase and epoxygenase products of arachidonic acid (AA) were elevated in all ICU groups compared with controls. sPLA2 predicted severity in COVID-19 and correlated with TxA2, LTE4 and the isoprostane, iPF2α-III, while PLA2G2D correlated with LTE4. The elevation in PGD2, like PGI2 and 12-HETE, exhibited relative specificity for COVID-19 and correlated with sPLA2 and the interleukin-13 receptor to drive lymphopenia, a marker of disease severity. Pro-inflammatory eicosanoids remained correlated with severity in COVID-19 28 days after admission. Amongst non-COVID ICU patients, elevations in 5- and 15-HETE and 9- and 13-HODE reflected viral rather than bacterial disease. Linoleic acid (LA) binds directly to SARS-CoV-2 and both LA and its di-HOME products reflected disease severity in COVID-19. In healthy marines, these lipids rose with seroconversion. Eicosanoids linked variably to the peripheral cellular immune response. PGE2, TxA2 and LTE4 correlated with T cell activation, as did PGD2 with non-B non-T cell activation. In COVID-19, LPS stimulated peripheral blood mononuclear cell PGF2α correlated with memory T cells, dendritic and NK cells while LA and DiHOMEs correlated with exhausted T cells. Three high abundance lipids - ChoE 18:3, LPC-O-16:0 and PC-O-30:0 - were altered specifically in COVID. LPC-O-16:0 was strongly correlated with T helper follicular cell activation and all three negatively correlated with multi-omic inflammatory pathways and disease severity. CONCLUSIONS A broad based lipidomic storm is a predictor of poor prognosis in ARDS. Alterations in sPLA2, PGD2 and 12-HETE and the high abundance lipids, ChoE 18:3, LPC-O-16:0 and PC-O-30:0 exhibit relative specificity for COVID-19 amongst such patients and correlate with the inflammatory response to link to disease severity.
Collapse
Affiliation(s)
- Hu Meng
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emanuela Ricciotti
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Antonijo Mrčela
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Divij Mathew
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Immunology and Immune HealthPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Liudmila L. Mazaleuskaya
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Soumita Ghosh
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Thomas G. Brooks
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Alexandra P. Turner
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Nicholas F. Lahens
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ai Wen Tan
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ashley Woolfork
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Greg Grant
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Katalin Susztak
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew G. Letizia
- Naval Medical Research CenterSilver SpringMarylandUSA
- Naval Medical Research Unit TWOSingaporeSingapore
| | - Stuart C. Sealfon
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - E. John Wherry
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Immunology and Immune HealthPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Critical CarePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Aalim M. Weljie
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Nuala J. Meyer
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Garret A. FitzGerald
- Institute for Translational Medicine and TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
23
|
Lv W, Mao H, Ruan Y, Li S, Shimizu K, Zhang L, Zhang C. Identification and immunological characterization of PLA2G2A and cell death-associated molecular clusters in idiopathic pulmonary fibrosis. Life Sci 2023; 331:122071. [PMID: 37673297 DOI: 10.1016/j.lfs.2023.122071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
AIMS Idiopathic pulmonary fibrosis (IPF) is a severe pulmonary interstitial pneumonia. Our study focuses on the role of PLA2 enzyme in the IPF to explore a more effective diagnosis and treatment mechanism of IPF. MAIN METHODS Transcriptome data of IPF from GEO database and bleomycin-induced pulmonary fibrosis mice were analyzed to identify PLA2 enzyme and their metabolite, lysophosphatidylcholines 18:0, in IPF. Based on PLA2G2A and PLA2G2D / PLA2G2A-associated cell death genes (PCDs), the consensus clustering analysis was used to identify the subtypes of IPF and the correlation between PLA2G2A and prognosis was analyzed. The machine learning (ML) models and artificial neural network (ANN) model was used to validate the diagnostic accuracy of PLA2s and PCDs in diagnosing IPF. The gene and protein expression of NLRP3, GSDMD, and CASP-1 was estimated in recombinant PLA2G2A protein induced MLE-12 cells. KEY FINDINGS The expression of PLA2G2D, PLA2G2A, and LPC18 significantly changed in IPF. Furtherly, PLA2G2A has a significant correlation with poor patient prognosis, which could predict the 2 or 3-years mortality rates of IPF. Two subtypes of IPF patients, identified based on PCDs, showed significant different immunoinfiltration. Recombinant PLA2G2A protein could induce the pyrotosis in the MLE-12 cell. The generalized linear model and ANN model of PLA2s or PCDs accurate diagnosis IPF. SIGNIFICANCE PLA2G2A is the most robustly associated gene with IPF among the PLA2s, which demonstrates a potential in diagnosing and prognostic value in IPF, and provides a foundation for further understanding and breakthroughs in IPF diagnosis and treatment.
Collapse
Affiliation(s)
- Weichao Lv
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hongcai Mao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Ruan
- Laboratory of Systematic Forest and Forest Products Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Shuaiyu Li
- Saigo Laboratory, School of Information Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Kuniyoshi Shimizu
- Laboratory of Systematic Forest and Forest Products Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Louqian Zhang
- Department of Thoratic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
24
|
Chaves-Filho AM, Braniff O, Angelova A, Deng Y, Tremblay MÈ. Chronic inflammation, neuroglial dysfunction, and plasmalogen deficiency as a new pathobiological hypothesis addressing the overlap between post-COVID-19 symptoms and myalgic encephalomyelitis/chronic fatigue syndrome. Brain Res Bull 2023; 201:110702. [PMID: 37423295 DOI: 10.1016/j.brainresbull.2023.110702] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/13/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
After five waves of coronavirus disease 2019 (COVID-19) outbreaks, it has been recognized that a significant portion of the affected individuals developed long-term debilitating symptoms marked by chronic fatigue, cognitive difficulties ("brain fog"), post-exertional malaise, and autonomic dysfunction. The onset, progression, and clinical presentation of this condition, generically named post-COVID-19 syndrome, overlap significantly with another enigmatic condition, referred to as myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Several pathobiological mechanisms have been proposed for ME/CFS, including redox imbalance, systemic and central nervous system inflammation, and mitochondrial dysfunction. Chronic inflammation and glial pathological reactivity are common hallmarks of several neurodegenerative and neuropsychiatric disorders and have been consistently associated with reduced central and peripheral levels of plasmalogens, one of the major phospholipid components of cell membranes with several homeostatic functions. Of great interest, recent evidence revealed a significant reduction of plasmalogen contents, biosynthesis, and metabolism in ME/CFS and acute COVID-19, with a strong association to symptom severity and other relevant clinical outcomes. These bioactive lipids have increasingly attracted attention due to their reduced levels representing a common pathophysiological manifestation between several disorders associated with aging and chronic inflammation. However, alterations in plasmalogen levels or their lipidic metabolism have not yet been examined in individuals suffering from post-COVID-19 symptoms. Here, we proposed a pathobiological model for post-COVID-19 and ME/CFS based on their common inflammation and dysfunctional glial reactivity, and highlighted the emerging implications of plasmalogen deficiency in the underlying mechanisms. Along with the promising outcomes of plasmalogen replacement therapy (PRT) for various neurodegenerative/neuropsychiatric disorders, we sought to propose PRT as a simple, effective, and safe strategy for the potential relief of the debilitating symptoms associated with ME/CFS and post-COVID-19 syndrome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, F-91400 Orsay, France
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec City, Québec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC) and Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
25
|
Murakami M, Sato H, Taketomi Y. Modulation of immunity by the secreted phospholipase A 2 family. Immunol Rev 2023; 317:42-70. [PMID: 37035998 DOI: 10.1111/imr.13205] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Among the phospholipase A2 (PLA2 ) superfamily, which typically catalyzes the sn-2 hydrolysis of phospholipids to yield fatty acids and lysophospholipids, the secreted PLA2 (sPLA2 ) family contains 11 isoforms in mammals. Individual sPLA2 s have unique enzymatic specificity toward fatty acids and polar heads of phospholipid substrates and display distinct tissue/cellular distributions, suggesting their distinct physiological functions. Recent studies using knockout and/or transgenic mice for a full set of sPLA2 s have revealed their roles in modulation of immunity and related disorders. Application of mass spectrometric lipidomics to these mice has enabled to identify target substrates and products of individual sPLA2 s in given tissue microenvironments. sPLA2 s hydrolyze not only phospholipids in the plasma membrane of activated, damaged or dying mammalian cells, but also extracellular phospholipids such as those in extracellular vesicles, microbe membranes, lipoproteins, surfactants, and dietary phospholipids, thereby exacerbating or ameliorating various diseases. The actions of sPLA2 s are dependent on, or independent of, the generation of fatty acid- or lysophospholipid-derived lipid mediators according to the pathophysiological contexts. In this review, we make an overview of our current understanding of the roles of individual sPLA2 s in various immune responses and associated diseases.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Hiroyasu Sato
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Alekseeva AS, Boldyrev IA. Alternative Targets for sPLA2 Activity: Role of Membrane-Enzyme Interactions. MEMBRANES 2023; 13:618. [PMID: 37504984 PMCID: PMC10384401 DOI: 10.3390/membranes13070618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023]
Abstract
The secreted phospholipases A2 (sPLA2s) play important roles both physiologically and pathologically, with their expression increasing significantly in diseases such as sepsis, inflammation, different cancers, glaucoma, obesity, Alzheimer's disease and even COVID-19. The fact has led to a large-scale search for inhibitors of these enzymes. In total, several dozen promising molecules have been proposed, but not a single one has successfully passed clinical trials. The failures in clinical studies motivated in-depth fundamental studies of PLA2s. Here we review alternative ways to control sPLA2 activity, outside its catalytic site. The concept can be realized by preventing sPLA2 from attaching to the membrane surface; by binding to an external protein which blocks sPLA2 hydrolytic activity; by preventing sPLA2 from orienting properly on the membrane surface; and by preventing substrate binding to the enzyme, keeping the catalytic site unaltered. Evidence in the literature is summarized in the review with the aim to serve as a starting point for new types of sPLA2 inhibitors.
Collapse
Affiliation(s)
- Anna S Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ivan A Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
27
|
Meng H, Sengupta A, Ricciotti E, Mrčela A, Mathew D, Mazaleuskaya LL, Ghosh S, Brooks TG, Turner AP, Schanoski AS, Lahens NF, Tan AW, Woolfork A, Grant G, Susztak K, Letizia AG, Sealfon SC, Wherry EJ, Laudanski K, Weljie AM, Meyer NB, FitzGerald GA. Deep Phenotyping of the Lipidomic Response in COVID and non-COVID Sepsis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543298. [PMID: 37398323 PMCID: PMC10312560 DOI: 10.1101/2023.06.02.543298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lipids may influence cellular penetrance by pathogens and the immune response that they evoke. Here we find a broad based lipidomic storm driven predominantly by secretory (s) phospholipase A 2 (sPLA 2 ) dependent eicosanoid production occurs in patients with sepsis of viral and bacterial origin and relates to disease severity in COVID-19. Elevations in the cyclooxygenase (COX) products of arachidonic acid (AA), PGD 2 and PGI 2 , and the AA lipoxygenase (LOX) product, 12-HETE, and a reduction in the high abundance lipids, ChoE 18:3, LPC-O-16:0 and PC-O-30:0 exhibit relative specificity for COVID-19 amongst such patients, correlate with the inflammatory response and link to disease severity. Linoleic acid (LA) binds directly to SARS-CoV-2 and both LA and its di-HOME products reflect disease severity in COVID-19. AA and LA metabolites and LPC-O-16:0 linked variably to the immune response. These studies yield prognostic biomarkers and therapeutic targets for patients with sepsis, including COVID-19. An interactive purpose built interactive network analysis tool was developed, allowing the community to interrogate connections across these multiomic data and generate novel hypotheses.
Collapse
|
28
|
Li D, Ding L, Luo J, Li QG. Prediction of mortality in pneumonia patients with connective tissue disease treated with glucocorticoids or/and immunosuppressants by machine learning. Front Immunol 2023; 14:1192369. [PMID: 37304293 PMCID: PMC10248221 DOI: 10.3389/fimmu.2023.1192369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Objectives The assessment of accurate mortality risk is essential for managing pneumonia patients with connective tissue disease (CTD) treated with glucocorticoids or/and immunosuppressants. This study aimed to construct a nomogram for predicting 90-day mortality in pneumonia patients using machine learning. Methods Data were obtained from the DRYAD database. Pneumonia patients with CTD were screened. The samples were randomly divided into a training cohort (70%) and a validation cohort (30%). A univariate Cox regression analysis was used to screen for prognostic variables in the training cohort. Prognostic variables were entered into the least absolute shrinkage and selection operator (Lasso) and a random survival forest (RSF) analysis was used to screen important prognostic variables. The overlapping prognostic variables of the two algorithms were entered into the stepwise Cox regression analysis to screen the main prognostic variables and construct a model. Model predictive power was assessed using the C-index, the calibration curve, and the clinical subgroup analysis (age, gender, interstitial lung disease, diabetes mellitus). The clinical benefits of the model were assessed using a decision curve analysis (DCA). Similarly, the C-index was calculated and the calibration curve was plotted to verify the model stability in the validation cohort. Results A total of 368 pneumonia patients with CTD (training cohort: 247; validation cohort: 121) treated with glucocorticoids or/and immunosuppressants were included. The univariate Cox regression analysis obtained 19 prognostic variables. Lasso and RSF algorithms obtained eight overlapping variables. The overlapping variables were entered into a stepwise Cox regression to obtain five variables (fever, cyanosis, blood urea nitrogen, ganciclovir treatment, and anti-pseudomonas treatment), and a prognostic model was constructed based on the five variables. The C-index of the construction nomogram of the training cohort was 0.808. The calibration curve, DCA results, and clinical subgroup analysis showed that the model also had good predictive power. Similarly, the C-index of the model in the validation cohort was 0.762 and the calibration curve had good predictive value. Conclusion In this study, the nomogram developed performed well in predicting the 90-day risk of death in pneumonia patients with CTD treated with glucocorticoids or/and immunosuppressants.
Collapse
Affiliation(s)
- Dongdong Li
- Medical College of Nanchang University, Nanchang, Jiangxi, China
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Liting Ding
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- JXHC Key Laboratory of Rheumatology and Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Jiao Luo
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qiu-Gen Li
- Medical College of Nanchang University, Nanchang, Jiangxi, China
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
29
|
Perumal R, Shunmugam L, Naidoo K, Abdool Karim SS, Wilkins D, Garzino-Demo A, Brechot C, Parthasarathy S, Vahlne A, Nikolich JŽ. Long COVID: a review and proposed visualization of the complexity of long COVID. Front Immunol 2023; 14:1117464. [PMID: 37153597 PMCID: PMC10157068 DOI: 10.3389/fimmu.2023.1117464] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
Post-Acute Sequelae of Severe Acute Respiratory Syndrome Coronavirus - 2 (SARS-CoV-2) infection, or Long COVID, is a prevailing second pandemic with nearly 100 million affected individuals globally and counting. We propose a visual description of the complexity of Long COVID and its pathogenesis that can be used by researchers, clinicians, and public health officials to guide the global effort toward an improved understanding of Long COVID and the eventual mechanism-based provision of care to afflicted patients. The proposed visualization or framework for Long COVID should be an evidence-based, dynamic, modular, and systems-level approach to the condition. Furthermore, with further research such a framework could establish the strength of the relationships between pre-existing conditions (or risk factors), biological mechanisms, and resulting clinical phenotypes and outcomes of Long COVID. Notwithstanding the significant contribution that disparities in access to care and social determinants of health have on outcomes and disease course of long COVID, our model focuses primarily on biological mechanisms. Accordingly, the proposed visualization sets out to guide scientific, clinical, and public health efforts to better understand and abrogate the health burden imposed by long COVID.
Collapse
Affiliation(s)
- Rubeshan Perumal
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), South African Medical Research Council (SAMRC) - CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
- Department of Pulmonology and Critical Care, Division of Internal Medicine, School Clinical Medicine, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
| | - Letitia Shunmugam
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), South African Medical Research Council (SAMRC) - CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), South African Medical Research Council (SAMRC) - CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Salim S. Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), South African Medical Research Council (SAMRC) - CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Durban, South Africa
| | - Dave Wilkins
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alfredo Garzino-Demo
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Christian Brechot
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
| | - Sairam Parthasarathy
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and University of Arizona College of Medicine-Tucson, Tucson, AZ, United States
| | - Anders Vahlne
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
- Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
| | - Janko Ž. Nikolich
- Long COVID Taskforce, The Global Virus Network, Baltimore, MD, United States
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, United States
- The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, United States
| |
Collapse
|
30
|
Farooqui AA, Farooqui T, Sun GY, Lin TN, Teh DBL, Ong WY. COVID-19, Blood Lipid Changes, and Thrombosis. Biomedicines 2023; 11:biomedicines11041181. [PMID: 37189799 DOI: 10.3390/biomedicines11041181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Although there is increasing evidence that oxidative stress and inflammation induced by COVID-19 may contribute to increased risk and severity of thromboses, the underlying mechanism(s) remain to be understood. The purpose of this review is to highlight the role of blood lipids in association with thrombosis events observed in COVID-19 patients. Among different types of phospholipases A2 that target cell membrane phospholipids, there is increasing focus on the inflammatory secretory phospholipase A2 IIA (sPLA2-IIA), which is associated with the severity of COVID-19. Analysis indicates increased sPLA2-IIA levels together with eicosanoids in the sera of COVID patients. sPLA2 could metabolise phospholipids in platelets, erythrocytes, and endothelial cells to produce arachidonic acid (ARA) and lysophospholipids. Arachidonic acid in platelets is metabolised to prostaglandin H2 and thromboxane A2, known for their pro-coagulation and vasoconstrictive properties. Lysophospholipids, such as lysophosphatidylcholine, could be metabolised by autotaxin (ATX) and further converted to lysophosphatidic acid (LPA). Increased ATX has been found in the serum of patients with COVID-19, and LPA has recently been found to induce NETosis, a clotting mechanism triggered by the release of extracellular fibres from neutrophils and a key feature of the COVID-19 hypercoagulable state. PLA2 could also catalyse the formation of platelet activating factor (PAF) from membrane ether phospholipids. Many of the above lipid mediators are increased in the blood of patients with COVID-19. Together, findings from analyses of blood lipids in COVID-19 patients suggest an important role for metabolites of sPLA2-IIA in COVID-19-associated coagulopathy (CAC).
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11929, Taiwan
| | - Daniel B L Teh
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119260, Singapore
- Neurobiology Research Programme, Life Sciences Institute, National University of Singapore, Singapore 119260, Singapore
| |
Collapse
|
31
|
Jaiswal A, Rehman R, Dutta J, Singh S, Ray A, Shridhar M, Jaisankar J, Bhatt M, Khandelwal D, Sahoo B, Ram A, Mabalirajan U. Cellular Distribution of Secreted Phospholipase A2 in Lungs of IPF Patients and Its Inhibition in Bleomycin-Induced Pulmonary Fibrosis in Mice. Cells 2023; 12:cells12071044. [PMID: 37048117 PMCID: PMC10092981 DOI: 10.3390/cells12071044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/13/2023] [Accepted: 01/29/2023] [Indexed: 04/03/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease with a very poor prognosis as it has a 2.5 to 5 years mean survival after proper diagnosis. Even nintedanib and pirfenidone cannot halt the progression, though they slow the progression of IPF. Hence, there is a need to understand the novel pathophysiology. Phospholipase A2 (PLA2) could be the ideal candidate to study in IPF, as they have a role in both inflammation and fibrosis. In the present study, we have shown the expression profile of various secretory Phospholipase A2 (PLA2) isoforms by analyzing publicly available transcriptome data of single cells from the lungs of healthy individuals and IPF patients. Among 11 members of sPLA2, PLA2G2A is found to be increased in the fibroblasts and mesothelial cells while PLA2G5 is found to be increased in the fibroblasts of IPF patients. We identified a subset of fibroblasts expressing high PLA2G2A with moderate expression of PLA2G5 and which are specific to IPF only; we named it as PLA2G2A+ IPF fibroblast. Pathway analysis revealed that these PLA2G2A+ IPF fibroblast have upregulation of both inflammatory and fibrosis-related pathways like the TGF-β signaling pathway, IL-17 signaling, the arachidonic acid metabolism pathway and ECM-receptor interaction. In addition to this, we found elevated levels of sPLA2-IIA in plasma samples of IPF patients in our cohort. PLA2G3, PLA2G10 and PLA2G12B are found in to be increased in certain epithelial cells of IPF patients. Thus, these findings indicate that these five isoforms have a disease-dominant role along with innate immune roles as these isoforms are found predominantly in structural cells of IPF patients. Further, we have targeted sPLA2 in mice model of bleomycin-induced lung fibrosis by pBPB, a known sPLA2 inhibitor. pBPB treatment attenuated lung fibrosis induced by bleomycin along with a reduction in TGF-β and deposition of extracellular matrix in lung. Thus, these findings indicate that these sPLA2 isoforms especially PLA2G2A may serve as a therapeutic target in lung fibrosis.
Collapse
|
32
|
Urazov S, Chernov A, Popov O, Klenkova N, Sushentseva N, Polkovnikova I, Apalko S, Kislyuk K, Pavlovich D, Ivanov A, Shcherbak S. Secretory Phospholipase A2 and Interleukin-6 Levels as Predictive Markers of the Severity and Outcome of Patients with COVID-19 Infections. Int J Mol Sci 2023; 24:5540. [PMID: 36982611 PMCID: PMC10059025 DOI: 10.3390/ijms24065540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Coronavirus disease (COVID-19) has become a global pandemic. COVID-19 patients need immediate diagnosis and rehabilitation, which makes it urgent to identify new protein markers for a prognosis of the severity and outcome of the disease. The aim of this study was to analyze the levels of interleukin-6 (IL-6) and secretory phospholipase (sPLA2) in the blood of patients regarding the severity and outcome of COVID-19 infection. The study included clinical and biochemical data obtained from 158 patients with COVID-19 treated at St. Petersburg City Hospital No. 40. A detailed clinical blood test was performed on all patients, as well as an assessment of IL-6, sPLA2, aspartate aminotransferase (AST), total protein, albumin, lactate dehydrogenase (LDH), APTT, fibrinogen, procalcitonin, D-dimer, C-reactive protein (CRB), ferritin, and glomerular filtration rate (GFR) levels. It was found that the levels of PLA2, IL-6, APTV, AST, CRP, LDH, IL-6, D-dimer, and ferritin, as well as the number of neutrophils, significantly increased in patients with mild to severe COVID-19 infections. The levels of IL-6 were positively correlated with APTT; the levels of AST, LDH, CRP, D-dimer, and ferritin; and the number of neutrophils. The increase in the level of sPLA2 was positively correlated with the levels of CRP, LDH, D-dimer, and ferritin, the number of neutrophils, and APTT, and negatively correlated with the levels of GFR and lymphocytes. High levels of IL-6 and PLA2 significantly increase the risk of a severe course by 13.7 and 2.24 times, and increase the risk of death from COVID-19 infection by 14.82 and 5.32 times, respectively. We have shown that the blood levels of sPLA2 and IL-6 increase in cases which eventually result in death and when patients are transferred to the ICU (as the severity of COVID-19 infection increases), showing that IL-6 and sPLA2 can be considered as early predictors of aggravation of COVID-19 infections.
Collapse
Affiliation(s)
- Stanislav Urazov
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Alexandr Chernov
- Bioenergetics Department of Life Sciences, Ben-Gurion University, Beer Sheva 84105, Israel
- Federal State Budgetary Institution of Science “Institute of Experimental Medicine”, 197376 St. Petersburg, Russia
| | - Oleg Popov
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Natalya Klenkova
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | | | | | - Svetlana Apalko
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | - Kseniya Kislyuk
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
| | | | - Andrey Ivanov
- Center for Clinical and Laboratory Diagnostics, Federal State Budgetary Military Educational Institution of Higher Education “Military Medical Academy named after S.M. Kirov”, 194044 St. Petersburg, Russia
| | - Sergey Shcherbak
- City Hospital 40 of Saint Petersburg, 197706 St. Petersburg, Russia
- Department of Postgraduate Medical Education, Federal State Budgetary Educational Institution of Higher Professional Education “Saint-Petersburg State University”, 199034 St. Petersburg, Russia
| |
Collapse
|
33
|
Nasopharyngeal SARS-CoV-2 load and perinatal outcomes after maternal infection diagnosed close to delivery. J Gynecol Obstet Hum Reprod 2023; 52:102569. [PMID: 36878392 PMCID: PMC9985537 DOI: 10.1016/j.jogoh.2023.102569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND The occurrence of COVID-19 during the pregnancy can cause several negative maternal and neonatal outcomes. Nasopharyngeal viral load is associated with inflammatory markers and might influence the disease severity in non-pregnant patients, but there are no data about the relationship between viral load and perinatal outcomes in pregnant patients. OBJECTIVE To investigate the hypothesis that nasopharyngeal SARS-CoV-2 load (estimated with real-time polymerase chain reaction delta cycle (ΔCt), measured in hospital clinical laboratories) is associated with perinatal outcomes, when COVID-19 is diagnosed in the third trimester of pregnancy. STUDY DESIGN International, retrospective, observational, multi-center, cohort study enrolling 390 women (393 neonates, three pairs of twins), analyzed with multivariate generalized linear models with skewed distributions (gamma) and identity link. The analyses were conducted for the whole population and then followed by a subgroup analysis according to the clinical severity of maternal COVID-19. RESULTS The estimated viral load in maternal nasopharynx is not significantly associated with gestational age at birth (adjusted B: -0.008 (95%CI: -0.04; 0.02); p = 0.889), birth weight (adjusted B: 4.29 (95%CI: -25; 35); p = 0.889), weight Z-score (adjusted B: -0.01 (95%CI: -0.03; 1); p = 0.336), 5' Apgar scores (adjusted B: -0. -9.8e-4 (95%CI: -0.01; 0.01); p = 0.889), prematurity (adjusted OR: -0.97 (95%CI: 0.93; 1.03); p = 0.766) and the small for gestational age status (adjusted OR: 1.03 (95%CI: 0.99; 1.07); p = 0.351). Similar results were obtained in subgroup analyses according to COVID-19 clinical severity. CONCLUSIONS The estimated maternal nasopharyngeal viral load in pregnant women affected by COVID-19 during the third trimester is not associated with main perinatal outcomes.
Collapse
|
34
|
Li X, Mei M, Pu X, Chen X, Li X, Meng F, He S, Li J, Gu W, Yang X, Zhang F, Yu J. Protective effect and mechanism of Polygonatum kingianum against hypoxia-induced injury. Heliyon 2023; 9:e14353. [PMID: 36967867 PMCID: PMC10034467 DOI: 10.1016/j.heliyon.2023.e14353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Background Hypoxia is an essential cause of fatigue and aging, and is associated with the occurrence and development of many diseases. Polygonatum kingianum (PK) is a deficiency-nourishing Chinese herbal medicine utilized as both medicine and food, and it has long been used to ameliorate human conditions associated with fatigue and aging over 2000 years in China. PK is an important genuine-medicinal-materials cultivated in Yunnan, China, and is used by the Bai, Wa, and Zhuang nationalities as a traditional medicine for enhancing immunity, anti-fatigue, and anti-aging, while the preventive effect of PK on hypoxia-induced injury and the underlying mechanism are indefinite. Aim of the study The present study aimed to evaluate the anti-hypoxia efficacy and understand the corresponding mechanism of PK water extract. Materials and methods The main active ingredients and targets of PK were predicted using network pharmacology, and the anti-hypoxia activities of Gracillin and Liquiritigenin were verified by in vitro experiments. The pharmacodynamic experiments were conducted to evaluate the major signal pathways of PK for detecting anti-hypoxia activity. Results Fifty active ingredients and 371 potential targets were screened by network pharmacology, then, we confirmed that Gracillin and Liquiritigenin were the main active components of PK to exert anti-hypoxia effect in vitro. The pharmacodynamic experiments revealed that PK enhanced the extension rate of the survival time (ERST) and regulated the targets-related biochemical parameters of rats under hypoxia, showing significant anti-hypoxia effects on rats. Conclusion The network pharmacology results suggested that PK exerts its anti-hypoxia effect through a multi-component and multi-target manner. Simultaneously, we also observed that Gracillin (saponins) and Liquiritigenin (flavonoids) are the main active components of PK to play a role in anti-hypoxia. The anti-hypoxia effect of PK could be associated with scavenging excess free radicals, maintaining the activities of antioxidant enzymes, and inhibiting oxidative stress due to lipid peroxidation. These findings provide insight into the Polygonatum kingianum as promising medicines or healthcare products for preventing and treating hypoxia.
Collapse
|
35
|
Jeican II, Inișca P, Gheban D, Anton V, Lazăr M, Vică ML, Mironescu D, Rebeleanu C, Crivii CB, Aluaș M, Albu S, Siserman CV. Histopathological Lung Findings in COVID-19 B.1.617.2 SARS-CoV-2 Delta Variant. J Pers Med 2023; 13:jpm13020279. [PMID: 36836513 PMCID: PMC9961426 DOI: 10.3390/jpm13020279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The Delta variant (Pango lineage B.1.617.2) is one of the most significant and aggressive variants of SARS-CoV-2. To the best of our knowledge, this is the first paper specifically studying pulmonary morphopathology in COVID-19 caused by the B.1.617.2 Delta variant. METHODS The study included 10 deceased patients (40-83 years) with the COVID-19 Delta variant. The necrotic lung fragments were obtained either by biopsy (six cases) or autopsy (four cases). Tissue samples were subjected to virology analysis for identification of the SARS-CoV-2 variant, histopathology, and immunohistochemistry (anti-SARS coronavirus mouse anti-virus antibody). RESULTS Virology analysis identified B.1.617.2 through genetic sequencing in eight cases, and in two cases, specific mutations of B.1.617.2 were identified. Macroscopically, in all autopsied cases, the lung had a particular appearance, purple in color, with increased consistency on palpation and abolished crepitations. Histopathologically, the most frequently observed lesions were acute pulmonary edema (70%) and diffuse alveolar damage at different stages. The immunohistochemical examination was positive for proteins of SARS-CoV-2 in 60% of cases on alveolocytes and in endothelial cells. CONCLUSIONS The histopathological lung findings in the B.1.617.2 Delta variant are similar to those previously described in COVID-19. Spike protein-binding antibodies were identified immunohistochemically both on alveolocytes and in the endothelial cells, showing the potential of indirect damage from thrombosis.
Collapse
Affiliation(s)
- Ionuț Isaia Jeican
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Patricia Inișca
- Department of Pathology, County Emergency Hospital Deva, 330084 Deva, Romania
| | - Dan Gheban
- Department of Pathology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Department of Pathology, Emergency Clinical Hospital for Children, 400370 Cluj-Napoca, Romania
| | - Vlad Anton
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Mihaela Lazăr
- Viral Respiratory Infections Laboratory, Cantacuzino National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Mihaela Laura Vică
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Cell and Molecular Biology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | | | - Codrin Rebeleanu
- Department of Legal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Carmen Bianca Crivii
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Maria Aluaș
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, Victor Babeș Str., No. 15, 400012 Cluj-Napoca, Romania
- Correspondence: (M.A.); (S.A.)
| | - Silviu Albu
- Department of Head and Neck Surgery and Otorhinolaryngology, University Clinical Hospital of Railway Company, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
- Correspondence: (M.A.); (S.A.)
| | - Costel Vasile Siserman
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Legal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| |
Collapse
|
36
|
Cardiovascular and Renal Comorbidities Included into Neural Networks Predict the Outcome in COVID-19 Patients Admitted to an Intensive Care Unit: Three-Center, Cross-Validation, Age- and Sex-Matched Study. J Cardiovasc Dev Dis 2023; 10:jcdd10020039. [PMID: 36826535 PMCID: PMC9967447 DOI: 10.3390/jcdd10020039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Here, we performed a multicenter, age- and sex-matched study to compare the efficiency of various machine learning algorithms in the prediction of COVID-19 fatal outcomes and to develop sensitive, specific, and robust artificial intelligence tools for the prompt triage of patients with severe COVID-19 in the intensive care unit setting. In a challenge against other established machine learning algorithms (decision trees, random forests, extra trees, neural networks, k-nearest neighbors, and gradient boosting: XGBoost, LightGBM, and CatBoost) and multivariate logistic regression as a reference, neural networks demonstrated the highest sensitivity, sufficient specificity, and excellent robustness. Further, neural networks based on coronary artery disease/chronic heart failure, stage 3-5 chronic kidney disease, blood urea nitrogen, and C-reactive protein as the predictors exceeded 90% sensitivity and 80% specificity, reaching AUROC of 0.866 at primary cross-validation and 0.849 at secondary cross-validation on virtual samples generated by the bootstrapping procedure. These results underscore the impact of cardiovascular and renal comorbidities in the context of thrombotic complications characteristic of severe COVID-19. As aforementioned predictors can be obtained from the case histories or are inexpensive to be measured at admission to the intensive care unit, we suggest this predictor composition is useful for the triage of critically ill COVID-19 patients.
Collapse
|
37
|
Russell P, Esser L, Hagemeyer CE, Voelcker NH. The potential impact of nanomedicine on COVID-19-induced thrombosis. NATURE NANOTECHNOLOGY 2023; 18:11-22. [PMID: 36536042 DOI: 10.1038/s41565-022-01270-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/12/2022] [Indexed: 06/17/2023]
Abstract
Extensive reports of pulmonary embolisms, ischaemic stroke and myocardial infarctions caused by coronavirus disease 2019 (COVID-19), as well as a significantly increased long-term risk of cardiovascular diseases in COVID-19 survivors, have highlighted severe deficiencies in our understanding of thromboinflammation and the need for new therapeutic options. Due to the complexity of the immunothrombosis pathophysiology, the efficacy of treatment with conventional anti-thrombotic medication is questioned. Thrombolytics do appear efficacious, but are hindered by severe bleeding risks, limiting their use. Nanomedicine can have profound impact in this context, protecting delicate (bio)pharmaceuticals from degradation en route and enabling delivery in a targeted and on demand manner. We provide an overview of the most promising nanocarrier systems and design strategies that may be adapted to develop nanomedicine for COVID-19-induced thromboinflammation, including dual-therapeutic approaches with antiviral and immunosuppressants. Resultant targeted and side-effect-free treatment may aid greatly in the fight against the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
- Peije Russell
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
38
|
Papadopoulos C, Spourita E, Tentes I, Steiropoulos P, Anagnostopoulos K. Red Blood Cell Malfunction in COVID-19: Molecular Mechanisms and Therapeutic Targets. Viral Immunol 2022; 35:649-652. [PMID: 36342758 DOI: 10.1089/vim.2021.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The world has been facing a pandemic for the past 2 years. COVID-19 still leads to millions of deaths worldwide, while deteriorating the global economy. The need for therapeutic targets, thus, remains. Interestingly, red blood cells, apart from gas exchange, also serve as modulators of innate and adaptive immunity. This function is accommodated mainly by surface molecules (proteins, lipids, and carbohydrates) and increased antioxidant capacity. However, under the circumstances of a disease state, red blood cells can become proinflammatory cells. Recent evidence has shown that, in the context of COVID-19, erythrocytes present protein oxidation, decreased antioxidant capacity, increased glycolysis, altered membrane lipidome, increased binding of Cytosine-Guanine (CpG) DNA and complement proteins, and low CD47 levels. These changes lead to an erythrocyte-dependent inflammation, which possibly participates in the hyperinflammation status of COVID-19. The current knowledge for the dysfunction of red blood cells during COVID-19 implies that the BAND3 protein and toll-like receptor 9 are potential therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eleftheria Spourita
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Tentes
- Laboratory of Biochemistry, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, University General Hospital Dragana, Alexandroupolis, Greece
| | | |
Collapse
|
39
|
Al-kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, Alorabi M, Hadi Al-Harcan NA, El-Bouseary MM, Batiha GES. Citicoline and COVID-19: vis-à-vis conjectured. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1463-1475. [PMID: 36063198 PMCID: PMC9442587 DOI: 10.1007/s00210-022-02284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a current pandemic disease caused by a novel severe acute respiratory syndrome coronavirus virus respiratory type 2 (SARS-CoV-2). SARS-CoV-2 infection is linked with various neurological manifestations due to cytokine-induced disruption of the blood brain barrier (BBB), neuroinflammation, and peripheral neuronal injury, or due to direct SARS-CoV-2 neurotropism. Of note, many repurposed agents were included in different therapeutic protocols in the management of COVID-19. These agents did not produce an effective therapeutic eradication of SARS-CoV-2, and continuing searching for novel anti-SARS-CoV-2 agents is a type of challenge nowadays. Therefore, this study aimed to review the potential anti-inflammatory and antioxidant effects of citicoline in the management of COVID-19.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali K. Al-Buhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Mohammed Alorabi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif, 21944 Saudi Arabia
| | - Nasser A. Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511 Egypt
| |
Collapse
|
40
|
Toelzer C, Gupta K, Yadav SKN, Hodgson L, Williamson MK, Buzas D, Borucu U, Powers K, Stenner R, Vasileiou K, Garzoni F, Fitzgerald D, Payré C, Gautam G, Lambeau G, Davidson AD, Verkade P, Frank M, Berger I, Schaffitzel C. The free fatty acid-binding pocket is a conserved hallmark in pathogenic β-coronavirus spike proteins from SARS-CoV to Omicron. SCIENCE ADVANCES 2022; 8:eadc9179. [PMID: 36417532 PMCID: PMC9683698 DOI: 10.1126/sciadv.adc9179] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/19/2022] [Indexed: 06/01/2023]
Abstract
As coronavirus disease 2019 (COVID-19) persists, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) emerge, accumulating spike (S) glycoprotein mutations. S receptor binding domain (RBD) comprises a free fatty acid (FFA)-binding pocket. FFA binding stabilizes a locked S conformation, interfering with virus infectivity. We provide evidence that the pocket is conserved in pathogenic β-coronaviruses (β-CoVs) infecting humans. SARS-CoV, MERS-CoV, SARS-CoV-2, and VOCs bind the essential FFA linoleic acid (LA), while binding is abolished by one mutation in common cold-causing HCoV-HKU1. In the SARS-CoV S structure, LA stabilizes the locked conformation, while the open, infectious conformation is devoid of LA. Electron tomography of SARS-CoV-2-infected cells reveals that LA treatment inhibits viral replication, resulting in fewer deformed virions. Our results establish FFA binding as a hallmark of pathogenic β-CoV infection and replication, setting the stage for FFA-based antiviral strategies to overcome COVID-19.
Collapse
Affiliation(s)
- Christine Toelzer
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Kapil Gupta
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
- Imophoron Ltd., St. Philips Central, Albert Rd, Bristol BS2 0XJ, UK
| | - Sathish K. N. Yadav
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Lorna Hodgson
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | | | - Dora Buzas
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
- Max Planck Bristol Centre for Minimal Biology, Cantock’s Close, Bristol BS8 1TS, UK
| | - Ufuk Borucu
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Kyle Powers
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Richard Stenner
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Kate Vasileiou
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Frederic Garzoni
- Imophoron Ltd., St. Philips Central, Albert Rd, Bristol BS2 0XJ, UK
| | - Daniel Fitzgerald
- Halo Therapeutics Ltd., St. Philips Central, Albert Rd, Bristol BS2 0XJ, UK
| | - Christine Payré
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Gunjan Gautam
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | - Gérard Lambeau
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Andrew D. Davidson
- Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Paul Verkade
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
| | | | - Imre Berger
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
- Max Planck Bristol Centre for Minimal Biology, Cantock’s Close, Bristol BS8 1TS, UK
- Halo Therapeutics Ltd., St. Philips Central, Albert Rd, Bristol BS2 0XJ, UK
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, UK
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, 24 Tyndall Ave, Bristol BS8 1TQ, UK
- Halo Therapeutics Ltd., St. Philips Central, Albert Rd, Bristol BS2 0XJ, UK
| |
Collapse
|
41
|
Lu E, Hara A, Sun S, Hallmark B, Snider JM, Seeds MC, Watkins JC, McCall CE, Zhang HH, Yao G, Chilton FH. Temporal Associations of Plasma Levels of the Secreted Phospholipase A 2 Family and Mortality in Severe COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.11.21.22282595. [PMID: 36451888 PMCID: PMC9709788 DOI: 10.1101/2022.11.21.22282595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Previous research suggests that group IIA secreted phospholipase A 2 (sPLA 2 -IIA) plays a role in and predicts severe COVID-19 disease. The current study reanalyzed a longitudinal proteomic data set to determine the temporal (days 0, 3 and 7) relationship between the levels of several members of a family of sPLA 2 isoforms and the severity of COVID-19 in 214 ICU patients. The levels of six secreted PLA 2 isoforms, sPLA 2 -IIA, sPLA 2 -V, sPLA 2 -X, sPLA 2 -IB, sPLA 2 -IIC, and sPLA 2 -XVI, increased over the first 7 ICU days in those who succumbed to the disease. sPLA 2 -IIA outperformed top ranked cytokines and chemokines as predictors of patient outcome. A decision tree corroborated these results with day 0 to day 3 kinetic changes of sPLA 2 -IIA that separated the death and severe categories from the mild category and increases from day 3 to day 7 significantly enriched the lethal category. In contrast, there was a time-dependent decrease in sPLA 2 -IID and sPLA 2 -XIIB in patients with severe or lethal disease, and these two isoforms were at higher levels in mild patients. Taken together, proteomic analysis revealed temporal sPLA 2 patterns that reflect the critical roles of sPLA 2 isoforms in severe COVID-19 disease.
Collapse
|
42
|
Schuller M, Oberhuber M, Prietl B, Zügner E, Prugger EM, Magnes C, Kirsch AH, Schmaldienst S, Pieber T, Brodmann M, Rosenkranz AR, Eller P, Eller K. Alterations in the Kynurenine-Tryptophan Pathway and Lipid Dysregulation Are Preserved Features of COVID-19 in Hemodialysis. Int J Mol Sci 2022; 23:ijms232214089. [PMID: 36430566 PMCID: PMC9698708 DOI: 10.3390/ijms232214089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19)-induced metabolic alterations have been proposed as a source for prognostic biomarkers and may harbor potential for therapeutic exploitation. However, the metabolic impact of COVID-19 in hemodialysis (HD), a setting of profound a priori alterations, remains unstudied. To evaluate potential COVID-19 biomarkers in end-stage kidney disease (CKD G5), we analyzed the plasma metabolites in different COVID-19 stages in patients with or without HD. We recruited 18 and 9 asymptomatic and mild, 11 and 11 moderate, 2 and 13 severely affected, and 10 and 6 uninfected HD and non-HD patients, respectively. Plasma samples were taken at the time of diagnosis and/or upon admission to the hospital and analyzed by targeted metabolomics and cytokine/chemokine profiling. Targeted metabolomics confirmed stage-dependent alterations of the metabolome in non-HD patients with COVID-19, which were less pronounced in HD patients. Elevated kynurenine levels and lipid dysregulation, shown by an increase in circulating free fatty acids and a decrease in lysophospholipids, could distinguish patients with moderate COVID-19 from non-infected individuals in both groups. Kynurenine and lipid alterations were also associated with ICAM-1 and IL-15 levels in HD and non-HD patients. Our findings support the kynurenine pathway and plasma lipids as universal biomarkers of moderate and severe COVID-19 independent of kidney function.
Collapse
Affiliation(s)
- Max Schuller
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Monika Oberhuber
- Center for Biomarker Research in Medicine, CBmed GmbH, 8010 Graz, Austria
| | - Barbara Prietl
- Center for Biomarker Research in Medicine, CBmed GmbH, 8010 Graz, Austria
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Elmar Zügner
- Institute for Biomedicine and Health Sciences (HEALTH), Joanneum Research Forschungsgesellschaft m.b.H., 8010 Graz, Austria
| | - Eva-Maria Prugger
- Institute for Biomedicine and Health Sciences (HEALTH), Joanneum Research Forschungsgesellschaft m.b.H., 8010 Graz, Austria
| | - Christoph Magnes
- Institute for Biomedicine and Health Sciences (HEALTH), Joanneum Research Forschungsgesellschaft m.b.H., 8010 Graz, Austria
| | - Alexander H. Kirsch
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | | | - Thomas Pieber
- Center for Biomarker Research in Medicine, CBmed GmbH, 8010 Graz, Austria
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Marianne Brodmann
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Alexander R. Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Kathrin Eller
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Correspondence: ; Tel.: +43-316-385-12170
| |
Collapse
|
43
|
Cardinali DP, Brown GM, Pandi-Perumal SR. Possible Application of Melatonin in Long COVID. Biomolecules 2022; 12:1646. [PMID: 36358996 PMCID: PMC9687267 DOI: 10.3390/biom12111646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 07/30/2023] Open
Abstract
Clinical sequelae and symptoms for a considerable number of COVID-19 patients can linger for months beyond the acute stage of SARS-CoV-2 infection, "long COVID". Among the long-term consequences of SARS-CoV-2 infection, cognitive issues (especially memory loss or "brain fog"), chronic fatigue, myalgia, and muscular weakness resembling myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are of importance. Melatonin may be particularly effective at reducing the signs and symptoms of SARS-CoV-2 infection due to its functions as an antioxidant, anti-inflammatory, and immuno-modulatory agent. Melatonin is also a chronobiotic medication effective in treating delirium and restoring the circadian imbalance seen in COVID patients in the intensive care unit. Additionally, as a cytoprotector, melatonin aids in the prevention of several COVID-19 comorbidities, including diabetes, metabolic syndrome, and ischemic and non-ischemic cardiovascular diseases. This narrative review discusses the application of melatonin as a neuroprotective agent to control cognitive deterioration ("brain fog") and pain in the ME/CFS syndrome-like documented in long COVID. Further studies on the therapeutic use of melatonin in the neurological sequelae of SARS-CoV-2 infection are warranted.
Collapse
Affiliation(s)
- Daniel P. Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires C1107AAZ, Argentina
| | - Gregory M. Brown
- Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Seithikurippu R. Pandi-Perumal
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| |
Collapse
|
44
|
He B, Zhan Y, Cai C, Yu D, Wei Q, Quan L, Huang D, Liu Y, Li Z, Liu L, Pan X. Common molecular mechanism and immune infiltration patterns of thoracic and abdominal aortic aneurysms. Front Immunol 2022; 13:1030976. [PMID: 36341412 PMCID: PMC9633949 DOI: 10.3389/fimmu.2022.1030976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 01/02/2024] Open
Abstract
BACKGROUND Aortic disease (aortic aneurysm (AA), dissection (AD)) is a serious threat to patient lives. Little is currently known about the molecular mechanisms and immune infiltration patterns underlying the development and progression of thoracic and abdominal aortic aneurysms (TAA and AAA), warranting further research. METHODS We downloaded AA (includes TAA and AAA) datasets from the GEO database. The potential biomarkers in TAA and AAA were identified using differential expression analysis and two machine-learning algorithms. The discrimination power of the potential biomarkers and their diagnostic accuracy was assessed in validation datasets using ROC curve analysis. Then, GSEA, KEGG, GO and DO analyses were conducted. Furthermore, two immuno-infiltration analysis algorithms were utilized to analyze the common immune infiltration patterns in TAA and AAA. Finally, a retrospective clinical study was performed on 78 patients with AD, and the serum from 6 patients was used for whole exome sequencing (WES). RESULTS The intersection of TAA and AAA datasets yielded 82 differentially expressed genes (DEGs). Subsequently, the biomarkers (CX3CR1 and HBB) were acquired by screening using two machine-learning algorithms and ROC curve analysis. The functional analysis of DEGs showed significant enrichment in inflammation and regulation of angiogenic pathways. Immune cell infiltration analysis revealed that adaptive and innate immune responses were closely linked to AA progression. However, neither CX3CR1 nor HBB was associated with B cell-mediated humoral immunity. CX3CR1 expression was correlated with macrophages and HBB with eosinophils. Finally, our retrospective clinical study revealed a hyperinflammatory environment in aortic disease. The WES study identified disease biomarkers and gene variants, some of which may be druggable. CONCLUSION The genes CX3CR1 and HBB can be used as common biomarkers in TAA and AAA. Large numbers of innate and adaptive immune cells are infiltrated in AA and are closely linked to the development and progression of AA. Moreover, CX3CR1 and HBB are highly correlated with the infiltration of immune cells and may be potential targets of immunotherapeutic drugs. Gene mutation research is a promising direction for the treatment of aortic disease.
Collapse
Affiliation(s)
- Bin He
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Ya Zhan
- The Third Hospital of MianYang, Sichuan Mental Health Center, MianYang, China
| | - Chunyu Cai
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Dianyou Yu
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Qinjiang Wei
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Liping Quan
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Da Huang
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yan Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zhile Li
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Li Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- College of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Xingshou Pan
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
45
|
Kurano M, Okamoto K, Jubishi D, Hashimoto H, Sakai E, Saigusa D, Kano K, Aoki J, Harada S, Okugawa S, Doi K, Moriya K, Yatomi Y. Dynamic modulations of sphingolipids and glycerophospholipids in COVID-19. Clin Transl Med 2022; 12:e1069. [PMID: 36214754 PMCID: PMC9549873 DOI: 10.1002/ctm2.1069] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A heterogeneous clinical phenotype is a characteristic of coronavirus disease 2019 (COVID-19). Therefore, investigating biomarkers associated with disease severity is important for understanding the mechanisms responsible for this heterogeneity and for developing novel agents to prevent critical conditions. This study aimed to elucidate the modulations of sphingolipids and glycerophospholipids, which have been shown to possess potent biological properties. METHODS We measured the serum sphingolipid and glycerophospholipid levels in a total of 887 samples from 215 COVID-19 subjects, plus 115 control subjects without infectious diseases and 109 subjects with infectious diseases other than COVID-19. RESULTS We observed the dynamic modulations of sphingolipids and glycerophospholipids in the serum of COVID-19 subjects, depending on the time course and severity. The elevation of C16:0 ceramide and lysophosphatidylinositol and decreases in C18:1 ceramide, dihydrosphingosine, lysophosphatidylglycerol, phosphatidylglycerol and phosphatidylinositol were specific to COVID-19. Regarding the association with maximum severity, phosphatidylinositol and phosphatidylcholine species with long unsaturated acyl chains were negatively associated, while lysophosphatidylethanolamine and phosphatidylethanolamine were positively associated with maximum severity during the early phase. Lysophosphatidylcholine and phosphatidylcholine had strong negative correlations with CRP, while phosphatidylethanolamine had strong positive ones. C16:0 ceramide, lysophosphatidylcholine, phosphatidylcholine and phosphatidylethanolamine species with long unsaturated acyl chains had negative correlations with D-dimer, while phosphatidylethanolamine species with short acyl chains and phosphatidylinositol had positive ones. Several species of phosphatidylcholine, phosphatidylethanolamine and sphingomyelin might serve as better biomarkers for predicting severe COVID-19 during the early phase than CRP and D-dimer. Compared with the lipid modulations seen in mice treated with lipopolysaccharide, tissue factor, or histone, the lipid modulations observed in severe COVID-19 were most akin to those in mice administered lipopolysaccharide. CONCLUSION A better understanding of the disturbances in sphingolipids and glycerophospholipids observed in this study will prompt further investigation to develop laboratory testing for predicting maximum severity and/or novel agents to suppress the aggravation of COVID-19.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory MedicineGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Koh Okamoto
- Department of Infectious DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Jubishi
- Department of Infectious DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Hideki Hashimoto
- Department of Infectious DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Eri Sakai
- Department of Clinical Laboratory MedicineGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical SciencesFaculty of Pharma‐ScienceTeikyo UniversityTokyoJapan
| | - Kuniyuki Kano
- Department of Health ChemistryGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Junken Aoki
- Department of Health ChemistryGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Sohei Harada
- Department of Infection Control and PreventionThe University of TokyoTokyoJapan
| | - Shu Okugawa
- Department of Infectious DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Kent Doi
- Department of Emergency and Critical Care MedicineThe University of Tokyo Hospital, Tokyo, Japan
| | - Kyoji Moriya
- Department of Infectious DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan,Department of Infection Control and PreventionThe University of TokyoTokyoJapan
| | - Yutaka Yatomi
- Department of Clinical Laboratory MedicineGraduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
46
|
Diversified Effects of COVID-19 as a Consequence of the Differential Metabolism of Phospholipids and Lipid Peroxidation Evaluated in the Plasma of Survivors and Deceased Patients upon Admission to the Hospital. Int J Mol Sci 2022; 23:ijms231911810. [PMID: 36233111 PMCID: PMC9570244 DOI: 10.3390/ijms231911810] [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: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
As a result of SARS-CoV-2 infection, inflammation develops, which promotes oxidative stress, leading to modification of phospholipid metabolism. Therefore, the aim of this study is to compare the effects of COVID-19 on the levels of phospholipid and free polyunsaturated fatty acids (PUFAs) and their metabolites produced in response to reactions with reactive oxygen species (ROS) and enzymes (cyclooxygenases-(COXs) and lipoxygenase-(LOX)) in the plasma of patients who either recovered or passed away within a week of hospitalization. In the plasma of COVID-19 patients, especially of the survivors, the actions of ROS and phospholipase A2 (PLA2) cause a decrease in phospholipid fatty acids level and an increase in free fatty acids (especially arachidonic acid) despite increased COXs and LOX activity. This is accompanied by an increased level in lipid peroxidation products (malondialdehyde and 8-isoprostaglandin F2α) and lipid mediators generated by enzymes. There is also an increase in eicosanoids, both pro-inflammatory as follows: thromboxane B2 and prostaglandin E2, and anti-inflammatory as follows: 15-deoxy-Δ-12,14-prostaglandin J2 and 12-hydroxyeicosatetraenoic acid, as well as endocannabinoids (anandamide-(AEA) and 2-arachidonylglycerol-(2-AG)) observed in the plasma of patients who recovered. Moreover, the expression of tumor necrosis factor α and interleukins (IL-6 and IL-10) is increased in patients who recovered. However, in the group of patients who died, elevated levels of N-oleoylethanolamine and N-palmitoylethanolamine are found. Since lipid mediators may have different functions depending on the onset of pathophysiological processes, a stronger pro-inflammatory response in patients who have recovered may be the result of the defensive response to SARS-CoV-2 in survivors associated with specific changes in the phospholipid metabolism, which could also be considered a prognostic factor.
Collapse
|
47
|
Mangini M, D’Angelo R, Vinciguerra C, Payré C, Lambeau G, Balestrieri B, Charles JF, Mariggiò S. Multimodal regulation of the osteoclastogenesis process by secreted group IIA phospholipase A 2. Front Cell Dev Biol 2022; 10:966950. [PMID: 36105351 PMCID: PMC9467450 DOI: 10.3389/fcell.2022.966950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023] Open
Abstract
Increasing evidence points to the involvement of group IIA secreted phospholipase A2 (sPLA2-IIA) in pathologies characterized by abnormal osteoclast bone-resorption activity. Here, the role of this moonlighting protein has been deepened in the osteoclastogenesis process driven by the RANKL cytokine in RAW264.7 macrophages and bone-marrow derived precursor cells from BALB/cJ mice. Inhibitors with distinct selectivity toward sPLA2-IIA activities and recombinant sPLA2-IIA (wild-type or catalytically inactive forms, full-length or partial protein sequences) were instrumental to dissect out sPLA2-IIA function, in conjunction with reduction of sPLA2-IIA expression using small-interfering-RNAs and precursor cells from Pla2g2a knock-out mice. The reported data indicate sPLA2-IIA participation in murine osteoclast maturation, control of syncytium formation and resorbing activity, by mechanisms that may be both catalytically dependent and independent. Of note, these studies provide a more complete understanding of the still enigmatic osteoclast multinucleation process, a crucial step for bone-resorbing activity, uncovering the role of sPLA2-IIA interaction with a still unidentified receptor to regulate osteoclast fusion through p38 SAPK activation. This could pave the way for the design of specific inhibitors of sPLA2-IIA binding to interacting partners implicated in osteoclast syncytium formation.
Collapse
Affiliation(s)
- Maria Mangini
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Rosa D’Angelo
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Caterina Vinciguerra
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Christine Payré
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne Sophia Antipolis, France
| | - Gérard Lambeau
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne Sophia Antipolis, France
| | - Barbara Balestrieri
- Jeff and Penny Vinik Center for Translational Immunology Research, Department of Medicine, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Julia F. Charles
- Departments of Orthopaedic Surgery and Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefania Mariggiò
- Institute of Protein Biochemistry, National Research Council, Naples, Italy,*Correspondence: Stefania Mariggiò,
| |
Collapse
|
48
|
Theodoropoulou MA, Koutoulogenis GS, Zhang L, Akrani I, Mikros E, Hilgenfeld R, Kokotos G. Identification of a Dual Inhibitor of Secreted Phospholipase A2 (GIIA sPLA2) and SARS-CoV-2 Main Protease. Pharmaceuticals (Basel) 2022; 15:ph15080961. [PMID: 36015109 PMCID: PMC9414318 DOI: 10.3390/ph15080961] [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: 04/04/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
The development of novel agents to combat COVID-19 is of high importance. SARS-CoV-2 main protease (Mpro) is a highly attractive target for the development of novel antivirals and a variety of inhibitors have already been developed. Accumulating evidence on the pathobiology of COVID-19 has shown that lipids and lipid metabolizing enzymes are critically involved in the severity of the infection. The purpose of the present study was to identify an inhibitor able to simultaneously inhibit both SARS-CoV-2 Mpro and phospholipase A2 (PLA2), an enzyme which plays a significant role in inflammatory diseases. Evaluating several PLA2 inhibitors, we demonstrate that the previously known potent inhibitor of Group IIA secretory PLA2, GK241, may also weakly inhibit SARS-CoV-2 Mpro. Molecular mechanics docking and molecular dynamics calculations shed light on the interactions between GK241 and SARS-CoV-2 Mpro. 2-Oxoamide GK241 may represent a lead molecular structure for the development of dual PLA2 and SARS-CoV-2 Mpro inhibitors.
Collapse
Affiliation(s)
- Maria A. Theodoropoulou
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (M.A.T.); (G.S.K.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Giorgos S. Koutoulogenis
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (M.A.T.); (G.S.K.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Linlin Zhang
- Institute of Molecular Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (L.Z.); (R.H.)
| | - Ifigeneia Akrani
- Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (I.A.); (E.M.)
| | - Emmanuel Mikros
- Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (I.A.); (E.M.)
- Athena Research and Innovation Center in Information Communication & Knowledge Technologies, 15125 Marousi, Greece
| | - Rolf Hilgenfeld
- Institute of Molecular Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; (L.Z.); (R.H.)
- German Center for Infection Research (DZIF), Hamburg–Lübeck–Borstel–Riems Site, University of Lübeck, 23562 Lübeck, Germany
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (M.A.T.); (G.S.K.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
- Correspondence: ; Tel.: +30-210-727-4462
| |
Collapse
|
49
|
Murakami M, Takamiya R, Miki Y, Sugimoto N, Nagasaki Y, Suzuki-Yamamoto T, Taketomi Y. Segregated functions of two cytosolic phospholipase A 2 isoforms (cPLA 2α and cPLA 2ε) in lipid mediator generation. Biochem Pharmacol 2022; 203:115176. [PMID: 35841927 DOI: 10.1016/j.bcp.2022.115176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/16/2022]
Abstract
Among the phospholipase A2 (PLA2) superfamily, group IVA cytosolic PLA2 (cPLA2α) is currently attracting much attention as a central regulator of arachidonic acid (AA) metabolism linked to eicosanoid biosynthesis. Following cell activation, cPLA2α selectively releases AA, a precursor of a variety of eicosanoids, from phospholipids in perinuclear membrane compartments. cPLA2α-null mice display various phenotypes that could be largely explained by reduced eicosanoid signaling. In contrast, group IVE cPLA2ε, another member of the cPLA2 family, acts as a Ca2+-dependent N-acyltransferase rather than a PLA2, thereby regulating the biosynthesis of N-acylethanolamines (NAEs), a unique class of lipid mediators with an anti-inflammatory effect. In response to Ca2+ signaling, cPLA2ε translocates to phosphatidylserine-rich organelle membranes in the endocytic/recycling pathway. In vivo, cPLA2ε is induced in keratinocytes of psoriatic skin, and its genetic deletion exacerbates psoriatic inflammation due to a marked reduction of NAE-related lipids. cPLA2ε also contributes to NAE generation in several if not all mouse tissues. Thus, the two members of the cPLA2 family, cPLA2α and cPLA2ε, catalyze distinct enzymatic reactions to mobilize distinct sets of lipid mediators, thereby differently regulating pathophysiological events in health and disease. Such segregation of the cPLA2α-eicosanoid and cPLA2ε-NAE pathways represents a new paradigm of research on PLA2s and lipid mediators.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Rina Takamiya
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Sugimoto
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Nagasaki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Nutritional Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Plasma Oxylipins and Their Precursors Are Strongly Associated with COVID-19 Severity and with Immune Response Markers. Metabolites 2022; 12:metabo12070619. [PMID: 35888743 PMCID: PMC9319897 DOI: 10.3390/metabo12070619] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is characterised by a dysregulated immune response, that involves signalling lipids acting as mediators of the inflammatory process along the innate and adaptive phases. To promote understanding of the disease biochemistry and provide targets for intervention, we applied a range of LC-MS platforms to analyse over 100 plasma samples from patients with varying COVID-19 severity and with detailed clinical information on inflammatory responses (>30 immune markers). The second publication in a series reports the results of quantitative LC-MS/MS profiling of 63 small lipids including oxylipins, free fatty acids, and endocannabinoids. Compared to samples taken from ward patients, intensive care unit (ICU) patients had 2−4-fold lower levels of arachidonic acid (AA) and its cyclooxygenase-derived prostanoids, as well as lipoxygenase derivatives, exhibiting negative correlations with inflammation markers. The same derivatives showed 2−5-fold increases in recovering ward patients, in paired comparison to early hospitalisation. In contrast, ICU patients showed elevated levels of oxylipins derived from poly-unsaturated fatty acids (PUFA) by non-enzymatic peroxidation or activity of soluble epoxide hydrolase (sEH), and these oxylipins positively correlated with markers of macrophage activation. The deficiency in AA enzymatic products and the lack of elevated intermediates of pro-resolving mediating lipids may result from the preference of alternative metabolic conversions rather than diminished stores of PUFA precursors. Supporting this, ICU patients showed 2-to-11-fold higher levels of linoleic acid (LA) and the corresponding fatty acyl glycerols of AA and LA, all strongly correlated with multiple markers of excessive immune response. Our results suggest that the altered oxylipin metabolism disrupts the expected shift from innate immune response to resolution of inflammation.
Collapse
|