1
|
Ferreira HB, Trindade F, Nogueira-Ferreira R, Leite-Moreira A, Ferreira R, Dias-Neto M, Domingues MR. Lipidomic insights on abdominal aortic aneurysm and peripheral arterial disease. J Mol Med (Berl) 2025; 103:365-380. [PMID: 40011252 PMCID: PMC12003574 DOI: 10.1007/s00109-025-02524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/10/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Abdominal aortic aneurysm (AAA) and peripheral arterial disease (PAD) are two cardiovascular diseases associated with considerable morbidity, mortality and quality of life impairment. As they are multifactorial diseases, several factors contribute to their pathogenesis, including oxidative stress and lipid peroxidation, and these may have key roles in the development of these pathologies. Alterations of the lipid metabolism and lipid profile have been reported in cardiovascular diseases but to a lesser extent in AAA and PAD. Modifications in the profile of some molecular lipid species, in particular, native phospholipid and triglyceride species were mainly reported for AAA, while alterations in the fatty acid profile were noticed in the case of PAD. Oxidized phospholipids were also reported for AAA. Although AAA and PAD have a common atherosclerotic root, lipidomics demonstrates the existence of distinct lipid. Lipidomic research regarding AAA and PAD is still scarce and should be set in motion to increase the knowledge on the lipid changes that occur in these diseases, contributing not only to the discovery of new biomarkers for diagnosis and prognosis assessment but also to tailor precision medicine in the clinical field.
Collapse
Affiliation(s)
- Helena Beatriz Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, 4200-319, Porto, Portugal
| | - Rita Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde São João, Porto, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
2
|
Carpanedo L, Rund KM, Wende LM, Kampschulte N, Schebb NH. LC-HRMS analysis of phospholipids bearing oxylipins. Anal Chim Acta 2024; 1326:343139. [PMID: 39260917 DOI: 10.1016/j.aca.2024.343139] [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: 05/16/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Several oxylipins including hydroxy- and epoxy-polyunsaturated fatty acids act as lipid mediators. In biological samples they can be present as non-esterified form, but the major part occurs esterified in phospholipids (PL) or other lipids. Esterified oxylipins are quantified indirectly after alkaline hydrolysis as non-esterified oxylipins. However, in this indirect analysis the information in which lipid class oxylipins are bound is lost. In this work, an untargeted liquid chromatography high-resolution mass spectrometry (LC-HRMS) method for the direct analysis of PL bearing oxylipins was developed. RESULTS Optimized reversed-phase LC separation achieved a sufficient separation of isobaric and isomeric PL from different lipid classes bearing oxylipin positional isomers. Individual PL species bearing oxylipins were identified based on retention time, precursor ion and characteristic product ions. The bound oxylipin could be characterized based on product ions resulting from the α-cleavage occurring at the hydroxy/epoxy group. PL sn-1/sn-2 isomers were identified based on the neutral loss of the fatty acyl in the sn-2 position. A total of 422 individual oxPL species from 7 different lipid classes i.e., PI, PS, PC, PE, PC-P, PC-O, and PE-P were detected in human serum and cells. This method enabled to determine in which PL class supplemented oxylipins are incorporated in HEK293 cells: 20:4;15OH, 20:4;14Ep, and 20:5;14Ep were mostly bound to PI. 20:4;8Ep and 20:5;8Ep were esterified to PC and PE while other oxylipins were mainly found in PC. SIGNIFICANCE The developed LC-HRMS method enables the comprehensive detection as well as the semi-quantification of isobaric and isomeric PL species bearing oxylipins. With this method, we show that the position of the oxidation has a great impact and directs the incorporation of oxylipins into the different PL classes in human cells.
Collapse
Affiliation(s)
- Laura Carpanedo
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Katharina M Rund
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Luca M Wende
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany.
| |
Collapse
|
3
|
Harsch BA, Borkowski K, Walker RE, Pedersen TL, Newman JW, Shearer GC. ABCA1 and apoA-I dependent 12-hydroxyeicosatetraenoic acid efflux regulates macrophage inflammatory signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603001. [PMID: 39026807 PMCID: PMC11257534 DOI: 10.1101/2024.07.11.603001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Aberrant high-density lipoprotein (HDL) function is implicated in inflammation-associated pathologies. While HDL ABCA1-mediated reverse cholesterol and phospholipid transport are well described, the movement of pro-/anti-inflammatory lipids has not been explored. HDL phospholipids are the largest reservoir of circulating arachidonic acid-derived oxylipins. Endotoxin-stimulation activates inflammatory cells leading to hydroxyeicosatetraenoic acid (HETE) production, oxylipins which are involved in inflammatory response coordination. Active signaling in the non-esterified (NE) pool is terminated by sequestration of HETEs as esterified (Es) forms and degradation. We speculate that an ABCA1-apoA-I-dependent efflux of HETEs from stimulated cells could regulate intracellular HETE availability. Here we test this hypothesis both in vitro and in vivo. In endotoxin-stimulated RAW-264.7 macrophages preloaded with d8-arachidonic acid we use compartmental tracer modeling to characterize the formation of HETEs, and their efflux into HDL. We found that in response to endotoxin: I) Cellular NE 12-HETE is positively associated with MCP-1 secretion (p<0.001); II) HETE transfer from NE to Es pools is ABCA1-depedent (p<0.001); III) Cellular Es HETEs are transported into media when both apoA-I and ABCA1 are present (p<0.001); IV) The stimulated efflux of HETEs >> arachidonate (p<0.001). Finally, in endotoxin challenged humans (n=17), we demonstrate that intravenous lipopolysaccharide (0.6 ng/kg body weight) resulted in accumulation of 12-HETE in HDL over a 168-hour follow-up. Therefore, HDL can suppress inflammatory responses in macrophages by regulating intracellular HETE content in an apoA-I/ABCA1 dependent manner. The described mechanism may apply to other oxylipins and explain anti-inflammatory properties of HDL. This newly defined HDL property opens new doors for the study of lipoprotein interactions in metabolic diseases.
Collapse
Affiliation(s)
- Brian A Harsch
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Kamil Borkowski
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA
| | - Rachel E Walker
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | | | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis CA
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis CA
- Department of Nutrition, University of California Davis, Davis CA
| | - Gregory C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
4
|
Hu G, Gu L, Wang R, Jian Q, Lv K, Xia M, Lai M, Shen T, Hu J, Yang S, Ye C, Zhang X, Wang Y, Xu X, Zhang F. Ethanolamine as a biomarker and biomarker-based therapy for diabetic retinopathy in glucose-well-controlled diabetic patients. Sci Bull (Beijing) 2024; 69:1920-1935. [PMID: 38423871 DOI: 10.1016/j.scib.2023.12.053] [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: 09/06/2023] [Revised: 11/21/2023] [Accepted: 12/29/2023] [Indexed: 03/02/2024]
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness among the working-age population. Although controlling blood glucose levels effectively reduces the incidence and development of DR to less than 50%, there are currently no diagnostic biomarkers or effective treatments for DR development in glucose-well-controlled diabetic patients (GW-DR). In this study, we established a prospective GW-DR cohort by strictly adhering to glycemic control guidelines and maintaining regular retinal examinations over a median 2-year follow-up period. The discovery cohort encompassed 71 individuals selected from a pool of 292 recruited diabetic patients at baseline, all of whom consistently maintained hemoglobin A1c (HbA1c) levels below 7% without experiencing hypoglycemia. Within this cohort of 71 individuals, 21 subsequently experienced new-onset GW-DR, resulting in an incidence rate of 29.6%. In the validation cohort, we also observed a significant GW-DR incidence rate of 17.9%. Employing targeted metabolomics, we investigated the metabolic characteristics of serum in GW-DR, revealing a significant association between lower levels of ethanolamine and GW-DR risk. This association was corroborated in the validation cohort, exhibiting superior diagnostic performance in distinguishing GW-DR from diabetes compared to the conventional risk factor HbA1c, with AUCs of 0.954 versus 0.506 and 0.906 versus 0.521 in the discovery and validation cohorts, respectively. Furthermore, in a streptozotocin (STZ)-induced diabetic rat model, ethanolamine attenuated diabetic retinal inflammation, accompanied by suppression of microglial diacylglycerol (DAG)-dependent protein kinase C (PKC) pathway activation. In conclusion, we propose that ethanolamine is a potential biomarker and represents a viable biomarker-based therapeutic option for GW-DR.
Collapse
Affiliation(s)
- Guangyi Hu
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Liping Gu
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ruonan Wang
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Qizhi Jian
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Kangjia Lv
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Mengxue Xia
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Mengyu Lai
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Tingting Shen
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jing Hu
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Sen Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Cunqi Ye
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaonan Zhang
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Xun Xu
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China.
| | - Fang Zhang
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Eye Institute of Shanghai Jiao Tong University School, Shanghai 200080, China; Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China.
| |
Collapse
|
5
|
Wang N, Yao T, Luo C, Sun L, Wang Y, Hou SX. Blockade of Arf1-mediated lipid metabolism in cancers promotes tumor infiltration of cytotoxic T cells via the LPE-PPARγ-NF-κB-CCL5 pathway. LIFE METABOLISM 2023; 2:load036. [PMID: 39872623 PMCID: PMC11749100 DOI: 10.1093/lifemeta/load036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 01/30/2025]
Abstract
Tumor immunotherapy has achieved breakthroughs in a variety of tumors. However, the systemic absence of T cells in tumors and immunosuppressive tumor microenvironment so far limits the efficacy of immunotherapy to a small population of patients. Therefore, novel agents to increase T-cell tumor infiltration are urgently needed in the clinic. We recently found that inhibition of the ADP-ribosylation factor 1 (Arf1)-mediated lipid metabolism not only kills cancer stem cells (CSCs) but also elicits an anti-tumor immune response. In this study, we revealed a mechanism that targeting Arf1 promotes the infiltration of cytotoxic T lymphocytes (CTLs) into tumors through the C-C chemokine ligand 5 (CCL5)- C-C chemokine receptor type 5 (CCR5) pathway. We found that blockage of Arf1 induces the production of the unsaturated fatty acid (PE 18:1) that binds and sequestrates peroxisome proliferator--activated receptor-γ (PPARγ) from the PPARγ-nuclear factor-κB (NF-κB) cytoplasmic complex. The released NF-κB was then phosphorylated and translocated into the nucleus to regulate the transcription of chemokine CCL5. CCL5 promoted infiltration of CTLs for tumor regression. Furthermore, the combination of the Arf1 inhibitor and programmed cell death protein 1 (PD-1) blockade induced an even stronger anti-tumor immunity. Therefore, targeting Arf1 represents a novel anti-tumor immune approach by provoking T-cell tumor infiltration and may provide a new strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Na Wang
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Tiange Yao
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Chenfei Luo
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Ling Sun
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Yuetong Wang
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Steven X Hou
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| |
Collapse
|
6
|
Abstract
Type 2 immunity mediates protective responses to helminths and pathological responses to allergens, but it also has broad roles in the maintenance of tissue integrity, including wound repair. Type 2 cytokines are known to promote fibrosis, an overzealous repair response, but their contribution to healthy wound repair is less well understood. This review discusses the evidence that the canonical type 2 cytokines, IL-4 and IL-13, are integral to the tissue repair process through two main pathways. First, essential for the progression of effective tissue repair, IL-4 and IL-13 suppress the initial inflammatory response to injury. Second, these cytokines regulate how the extracellular matrix is modified, broken down, and rebuilt for effective repair. IL-4 and/or IL-13 amplifies multiple aspects of the tissue repair response, but many of these pathways are highly redundant and can be induced by other signals. Therefore, the exact contribution of IL-4Rα signaling remains difficult to unravel.
Collapse
Affiliation(s)
- Judith E Allen
- Lydia Becker Institute for Immunology and Inflammation and Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
7
|
Compartmentalized regulation of lipid signaling in oxidative stress and inflammation: Plasmalogens, oxidized lipids and ferroptosis as new paradigms of bioactive lipid research. Prog Lipid Res 2023; 89:101207. [PMID: 36464139 DOI: 10.1016/j.plipres.2022.101207] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Perturbations in lipid homeostasis combined with conditions favoring oxidative stress constitute a hallmark of the inflammatory response. In this review we focus on the most recent results concerning lipid signaling in various oxidative stress-mediated responses and inflammation. These include phagocytosis and ferroptosis. The best characterized event, common to these responses, is the synthesis of oxygenated metabolites of arachidonic acid and other polyunsaturated fatty acids. Major developments in this area have highlighted the importance of compartmentalization of the enzymes and lipid substrates in shaping the appropriate response. In parallel, other relevant lipid metabolic pathways are also activated and, until recently, there has been a general lack of knowledge on the enzyme regulation and molecular mechanisms operating in these pathways. Specifically, data accumulated in recent years on the regulation and biological significance of plasmalogens and oxidized phospholipids have expanded our knowledge on the involvement of lipid metabolism in the progression of disease and the return to homeostasis. These recent major developments have helped to establish the concept of membrane phospholipids as cellular repositories for the compartmentalized production of bioactive lipids involved in cellular regulation. Importantly, an enzyme classically described as being involved in regulating the homeostatic turnover of phospholipids, namely the group VIA Ca2+-independent phospholipase A2 (iPLA2β), has taken center stage in oxidative stress and inflammation research owing to its key involvement in regulating metabolic and ferroptotic signals arising from membrane phospholipids. Understanding the role of iPLA2β in ferroptosis and metabolism not only broadens our knowledge of disease but also opens possible new horizons for this enzyme as a target for therapeutic intervention.
Collapse
|
8
|
Millrine D, Rice CM, Fernandez JU, Jones SA. Tracking the Host Response to Infection in Peritoneal Models of Acute Resolving Inflammation. Methods Mol Biol 2023; 2691:81-95. [PMID: 37355539 DOI: 10.1007/978-1-0716-3331-1_7] [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] [Indexed: 06/26/2023]
Abstract
Antimicrobial host defense is dependent on the rapid recruitment of inflammatory cells to the site of infection, the elimination of invading pathogens, and the efficient resolution of inflammation that minimizes damage to the host. The peritoneal cavity provides an accessible and physiologically relevant system where the delicate balance of these processes may be studied. Here, we describe murine models of peritoneal inflammation that enable studies of competent antimicrobial immunity and inflammation-associated tissue damage as a consequence of recurrent bacterial challenge. The inflammatory hallmarks of these models reflect the clinical and molecular features of peritonitis seen in renal failure patients on peritoneal dialysis. The development of these models relies on the preparation of a cell-free supernatant derived from an isolate of Staphylococcus epidermidis (termed SES). Intraperitoneal administration of SES induces a Toll-like receptor 2-driven acute inflammatory response that is characterized by an initial transient influx of neutrophils that are replaced by a more sustained recruitment of mononuclear cells and lymphocytes. Adaptation of this model using a repeated administration of SES allows investigations into the development of adaptive immunity and the hallmarks associated with tissue remodelling and fibrosis. These models are therefore clinically relevant and provide exciting opportunities to study innate and adaptive immunity and the response of the stromal tissue compartment to bacterial infection and the ensuing inflammatory reaction.
Collapse
Affiliation(s)
- David Millrine
- Medical Research Council Protein Phosphorylation & Ubiquitylation Unit (MRC-PPU), School of Life Sciences, University of Dundee, Dundee, UK
| | - Christopher M Rice
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | - Javier U Fernandez
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK
- Systems Immunity University Research Institute, Cardiff University, Cardiff, Wales, UK
| | - Simon A Jones
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK.
- Systems Immunity University Research Institute, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
9
|
Kuksis A, Pruzanski W. Hydrolysis of polyhydroxy polyunsaturated fatty acid-glycerophosphocholines by Group IIA, V, and X secretory phospholipases A 2. Lipids 2023; 58:3-17. [PMID: 36114729 DOI: 10.1002/lipd.12359] [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: 05/23/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 02/04/2023]
Abstract
It is widely accepted that unesterified polyunsaturated ω-6 and ω-3 fatty acids (PUFA) are converted through various lipoxygenases, cyclooxygenases, and cytochrome P450 enzymes to a range of oxygenated derivatives (oxylipins), among which the polyhydroxides of unesterified PUFA have recently been recognized as cell signaling molecules with anti-inflammatory and pro-resolving properties, known as specialized pro-resolving mediators (SPMs). This study investigates the mono-, di-, and trihydroxy 16:0/PUFA-GPCs, and the corresponding 16:0/SPM-GPC, in plasma lipoproteins. We describe the isolation and identification of mono-, di-, and trihydroxy AA, EPA, and DHA-GPC in plasma LDL, HDL, HDL3, and acute phase HDL using normal phase LC/ESI-MS, as previously reported. The lipoproteins contained variable amounts of the polyhydroxy-PUFA-GPC (0-10 nmol/mg protein), likely the product of lipid peroxidation and the action of various lipoxygenases and cytochrome P450 enzymes on both free fatty acids and the parent GPCs. Polyhydroxy-PUFA-GPC was hydrolyzed to variable extent (20%-80%) by the different secretory phospholipases A2 (sPLA2 s), with Group IIA sPLA2 showing the lowest and Group X sPLA2 the highest activity. Surprisingly, the trihydroxy-16:0/PUFA-GPC of APHDL was largely absent, while large amounts of unidentified material had migrated in the free fatty acid elution area. The free fatty acid mass spectra were consistent with that anticipated for branched chain polyhydroxy fatty acids. There was general agreement between the masses determined by LC/ESI-MS for the polyhydroxy PUFA-GPC and the masses calculated for the GPC equivalents of resolvins, protectins, and maresins using the fatty acid structures reported in the literature.
Collapse
Affiliation(s)
- Arnis Kuksis
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | | |
Collapse
|
10
|
Gong C, Ji Q, Wu M, Tu Z, Lei K, Luo M, Liu J, Lin L, Li K, Li J, Huang K, Zhu X. Ferroptosis in tumor immunity and therapy. J Cell Mol Med 2022; 26:5565-5579. [DOI: 10.1111/jcmm.17529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Chuandong Gong
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Qiankun Ji
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Miaojing Wu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Zewei Tu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Kunjian Lei
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Min Luo
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Junzhe Liu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Li Lin
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Kuangxun Li
- College of Queen Mary Nanchang University Nanchang China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit Second Affiliated Hospital of Nanchang University Nanchang China
| | - Kai Huang
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| | - Xingen Zhu
- Department of Neurosurgery The Second Affiliated Hospital of Nanchang University Nanchang China
- Institute of Neuroscience, Nanchang University Nanchang China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases Nanchang China
| |
Collapse
|
11
|
Petkevicius K, Palmgren H, Glover MS, Ahnmark A, Andréasson AC, Madeyski-Bengtson K, Kawana H, Allman EL, Kaper D, Uhrbom M, Andersson L, Aasehaug L, Forsström J, Wallin S, Ahlstedt I, Leke R, Karlsson D, González-King H, Löfgren L, Nilsson R, Pellegrini G, Kono N, Aoki J, Hess S, Sienski G, Pilon M, Bohlooly-Y M, Maresca M, Peng XR. TLCD1 and TLCD2 regulate cellular phosphatidylethanolamine composition and promote the progression of non-alcoholic steatohepatitis. Nat Commun 2022; 13:6020. [PMID: 36241646 PMCID: PMC9568529 DOI: 10.1038/s41467-022-33735-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
The fatty acid composition of phosphatidylethanolamine (PE) determines cellular metabolism, oxidative stress, and inflammation. However, our understanding of how cells regulate PE composition is limited. Here, we identify a genetic locus on mouse chromosome 11, containing two poorly characterized genes Tlcd1 and Tlcd2, that strongly influences PE composition. We generated Tlcd1/2 double-knockout (DKO) mice and found that they have reduced levels of hepatic monounsaturated fatty acid (MUFA)-containing PE species. Mechanistically, TLCD1/2 proteins act cell intrinsically to promote the incorporation of MUFAs into PEs. Furthermore, TLCD1/2 interact with the mitochondria in an evolutionarily conserved manner and regulate mitochondrial PE composition. Lastly, we demonstrate the biological relevance of our findings in dietary models of metabolic disease, where Tlcd1/2 DKO mice display attenuated development of non-alcoholic steatohepatitis compared to controls. Overall, we identify TLCD1/2 proteins as key regulators of cellular PE composition, with our findings having broad implications in understanding and treating disease. The regulation of cellular phosphatidylethanolamine (PE) acyl chain composition is poorly understood. Here, the authors show that TLCD1 and TLCD2 proteins mediate the formation of monounsaturated fatty acid-containing PE species and promote the progression of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Kasparas Petkevicius
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden. .,Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Henrik Palmgren
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Matthew S Glover
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrea Ahnmark
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anne-Christine Andréasson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Tokyo, Japan
| | - Erik L Allman
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Delaney Kaper
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Martin Uhrbom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Liselotte Andersson
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Leif Aasehaug
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Johan Forsström
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Simonetta Wallin
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ingela Ahlstedt
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Renata Leke
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Karlsson
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hernán González-King
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Löfgren
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ralf Nilsson
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Tokyo, Japan
| | - Sonja Hess
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Grzegorz Sienski
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Marcello Maresca
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
12
|
Petan T, Manček-Keber M. Half is enough: Oxidized lysophospholipids as novel bioactive molecules. Free Radic Biol Med 2022; 188:351-362. [PMID: 35779690 DOI: 10.1016/j.freeradbiomed.2022.06.228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Studies in the last decade have established the roles of oxidized phospholipids as modulators of various cellular processes, from inflammation and immunity to cell death. Oxidized lysophospholipids, formed through the activity of phospholipases and oxidative enzymes and lacking an acyl chain in comparison with parent phospholipids, are now emerging as novel bioactive lipid mediators. Their detection and structural characterization have been limited in the past due to low amounts and the complexity of their biosynthetic and removal pathways, but recent studies have unequivocally demonstrated their formation under inflammatory conditions. The involvement of oxidized lysophospholipids in immune regulation classifies them as damage-associated molecular patterns (DAMPs), which can promote sterile inflammation and contribute to autoimmune and chronic diseases as well as aging-related diseases. Their signaling pathways are just beginning to be revealed. As the first publications indicate that oxidized lysophospholipids use the same receptors as pathogen-associated molecular patterns (PAMPs), it is likely that the inhibition of signaling pathways activated by oxidized lysophospholipids would affect innate immunity per se. On the other hand, inhibition or modulation of their enzymatic formation, which would not interfere with the response to pathogens, might be beneficial and is potentially a promising new field of research.
Collapse
Affiliation(s)
- Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, 1000, Ljubljana, Slovenia.
| | - Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia.
| |
Collapse
|
13
|
Abedi M, Rahgozar S. Puzzling Out Iron Complications in Cancer Drug Resistance. Crit Rev Oncol Hematol 2022; 178:103772. [PMID: 35914667 DOI: 10.1016/j.critrevonc.2022.103772] [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: 05/28/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/09/2022] Open
Abstract
Iron metabolism are frequently disrupted in cancer. Patients with cancer are prone to anemia and receive transfusions frequently; the condition which results in iron overload, contributing to serious therapeutic complications. Iron is introduced as a carcinogen that may increase tumor growth. However, investigations regarding its impact on response to chemotherapy, particularly the induction of drug resistance are still limited. Here, iron contribution to cell signaling and various molecular mechanisms underlying iron-mediated drug resistance are described. A dual role of this vital element in cancer treatment is also addressed. On one hand, the need to administer iron chelators to surmount iron overload and improve the sensitivity of tumor cells to chemotherapy is discussed. On the other hand, the necessary application of iron as a therapeutic option by iron-oxide nanoparticles or ferroptosis inducers is explained. Authors hope that this paper can help unravel the clinical complications related to iron in cancer therapy.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
14
|
Gao W, Wang X, Zhou Y, Wang X, Yu Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther 2022; 7:196. [PMID: 35725836 PMCID: PMC9208265 DOI: 10.1038/s41392-022-01046-3] [Citation(s) in RCA: 494] [Impact Index Per Article: 164.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, immunotherapy represented by immune checkpoint inhibitors (ICIs) has led to unprecedented breakthroughs in cancer treatment. However, the fact that many tumors respond poorly or even not to ICIs, partly caused by the absence of tumor-infiltrating lymphocytes (TILs), significantly limits the application of ICIs. Converting these immune “cold” tumors into “hot” tumors that may respond to ICIs is an unsolved question in cancer immunotherapy. Since it is a general characteristic of cancers to resist apoptosis, induction of non-apoptotic regulated cell death (RCD) is emerging as a new cancer treatment strategy. Recently, several studies have revealed the interaction between non-apoptotic RCD and antitumor immunity. Specifically, autophagy, ferroptosis, pyroptosis, and necroptosis exhibit synergistic antitumor immune responses while possibly exerting inhibitory effects on antitumor immune responses. Thus, targeted therapies (inducers or inhibitors) against autophagy, ferroptosis, pyroptosis, and necroptosis in combination with immunotherapy may exert potent antitumor activity, even in tumors resistant to ICIs. This review summarizes the multilevel relationship between antitumor immunity and non-apoptotic RCD, including autophagy, ferroptosis, pyroptosis, and necroptosis, and the potential targeting application of non-apoptotic RCD to improve the efficacy of immunotherapy in malignancy.
Collapse
Affiliation(s)
- Weitong Gao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, changsha, 410008, China
| | - Yang Zhou
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xueqian Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yan Yu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
15
|
Protty MB, Jenkins PV, Collins PW, O'Donnell VB. The role of procoagulant phospholipids on the surface of circulating blood cells in thrombosis and haemostasis. Open Biol 2022; 12:210318. [PMID: 35440201 PMCID: PMC9019515 DOI: 10.1098/rsob.210318] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Phospholipids (PLs) are found in all cell types and are required for structural support and cell activation signalling pathways. In resting cells, PLs are asymmetrically distributed throughout the plasma membrane with native procoagulant aminophospholipids (aPLs) being actively maintained in the inner leaflet of the membrane. Upon platelet activation, aPLs rapidly externalize to the outer leaflet and are essential for supporting the coagulation cascade by providing binding sites for factors in the cell-based model. More recent work has uncovered a role for enzymatically oxidized PLs (eoxPLs) in facilitating coagulation, working in concert with native aPLs. Despite this, the role of aPLs and eoxPLs in thrombo-inflammatory conditions, such as arterial and venous thrombosis, has not been fully elucidated. In this review, we describe the biochemical structures, distribution and regulation of aPL externalization and summarize the literature on eoxPL generation in circulating blood cells. We focus on the currently understood role of these lipids in mediating coagulation reactions in vitro, in vivo and in human thrombotic disease. Finally, we highlight gaps in our understanding in how these lipids vary in health and disease, which may place them as future therapeutic targets for the management of thrombo-inflammatory conditions.
Collapse
Affiliation(s)
- Majd B. Protty
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - P. Vince Jenkins
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Peter W. Collins
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | | |
Collapse
|
16
|
Involvement of Proinflammatory Arachidonic Acid (ARA) Derivatives in Crohn’s Disease (CD) and Ulcerative Colitis (UC). J Clin Med 2022; 11:jcm11071861. [PMID: 35407469 PMCID: PMC8999554 DOI: 10.3390/jcm11071861] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/10/2022] Open
Abstract
Recently, an increase in the incidence of inflammatory bowel disease (IBD) has been observed, especially among children and adolescents. Currently, few studies focus on the differentiation of inflammation in IBD subunits, i.e., Crohn’s Disease (CD) and Ulcerative Colitis (UC). The aim of this study was to compare the concentrations of proinflammatory mediators of arachidonic acid (ARA) and linoleic acid (LA) in patients with CD (n = 34) and UC (n = 30), in order to identify differences in inflammation in both diseases and within the same entity, according to disease activity. Sixty-four adolescents with a mean age of 13.76 ± 2.69 and 14.15 ± 3.31, for CD and UC, respectively, were enrolled in the study. Biochemical analysis of ARA and LA derivatives was performed using a liquid chromatography. A trend was observed in the concentration of 15S-HETE (hydroxyeicosatetraenoic acids) in CD relative to UC. The active phase of both diseases showed a higher 15S-HETE concentration in active CD relative to active UC. Comparing patients with CD with active and inactive disease showed a trend of increased levels of thromboxane B2, leukotriene B4 and 9S-HODE (hydroxyoctadecadienoic acid) in the active versus the inactive disease. We also observed statistically significantly higher levels of 12S-HETE in inactive CD relative to active CD. In the UC group, on the other hand, statistically significantly higher levels of prostaglandin E2 and 16RS-HETE were observed in active UC relative to inactive UC. Moreover, significantly higher concentrations of LTX A4 5S, 6R were observed in inactive UC relative to the active phase. In conclusion, the present study indicated the activity of the 15-LOX pathway in CD. Further studies involving lipid mediators in patients with IBD may contribute to the development of new therapies for the treatment of IBD. The identification of differences in the course of inflammation may help to target therapy in CD and UC, and perhaps allow the introduction of an additional diagnostic marker between the two main IBD subtypes.
Collapse
|
17
|
O'Donnell VB. New appreciation for an old pathway: the Lands Cycle moves into new arenas in health and disease. Biochem Soc Trans 2022; 50:1-11. [PMID: 35225335 PMCID: PMC9022965 DOI: 10.1042/bst20210579] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 02/08/2023]
Abstract
The Lands Pathway is a fundamental biochemical process named for its discovery by William EM Lands and revealed in a series of seminal papers published in the Journal of Biological Chemistry between 1958-65. It describes the selective placement in phospholipids of acyl chains, by phospholipid acyltransferases. This pathway has formed a core component of our knowledge of phospholipid and also diglyceride metabolism in mammalian tissues for over 60 years now. Our understanding of how the Lands pathways are enzymatically mediated via large families of related gene products that display both substrate and tissue specificity has grown exponentially since. Recent studies building on this are starting to reveal key roles for the Lands pathway in specific scenarios, in particular inflammation, immunity and inflammation. This review will cover the Lands cycle from historical perspectives first, then present new information on how this important cycle forms a central regulatory node connecting fatty acyl and phospholipid metabolism and how its altered regulation may present new opportunities for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Valerie B. O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4SN, U.K
| |
Collapse
|
18
|
Aoyagi R, Yamamoto T, Furukawa Y, Arita M. Characterization of the Structural Diversity and Structure-Specific Behavior of Oxidized Phospholipids by LC-MS/MS. Chem Pharm Bull (Tokyo) 2021; 69:953-961. [PMID: 34602576 DOI: 10.1248/cpb.c21-00274] [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: 11/22/2022]
Abstract
Polyunsaturated fatty acids (PUFAs), esterified to phospholipids, are susceptible to oxidation. They form oxidized phospholipids (OxPLs) by oxygenases or reactive oxygen species (ROS), or both. These OxPLs are associated with various diseases, such as atherosclerosis, pulmonary injuries, neurodegenerative diseases, cancer, and diabetes. Since many types of OxPLs seem to be generated in vivo, precise determination of their structural diversity is required to understand their potential structure-specific functions. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is a powerful method to quantitatively measure the structural diversity of OxPLs present in biological samples. This review outlines recent advances in analytical methods for OxPLs and their physiological relevance in health and diseases.
Collapse
Affiliation(s)
- Ryohei Aoyagi
- Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS)
| | - Takahiro Yamamoto
- Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS)
| | - Yuuki Furukawa
- Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS)
| | - Makoto Arita
- Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS).,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama-City University
| |
Collapse
|
19
|
Liu X, Sims HF, Jenkins CM, Guan S, Dilthey BG, Gross RW. 12-LOX catalyzes the oxidation of 2-arachidonoyl-lysolipids in platelets generating eicosanoid-lysolipids that are attenuated by iPLA 2γ knockout. J Biol Chem 2020; 295:5307-5320. [PMID: 32161117 PMCID: PMC7170522 DOI: 10.1074/jbc.ra119.012296] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
The canonical pathway of eicosanoid production in most mammalian cells is initiated by phospholipase A2-mediated release of arachidonic acid, followed by its enzymatic oxidation resulting in a vast array of eicosanoid products. However, recent work has demonstrated that the major phospholipase in mitochondria, iPLA2γ (patatin-like phospholipase domain containing 8 (PNPLA8)), possesses sn-1 specificity, with polyunsaturated fatty acids at the sn-2 position generating polyunsaturated sn-2-acyl lysophospholipids. Through strategic chemical derivatization, chiral chromatographic separation, and multistage tandem MS, here we first demonstrate that human platelet-type 12-lipoxygenase (12-LOX) can directly catalyze the regioselective and stereospecific oxidation of 2-arachidonoyl-lysophosphatidylcholine (2-AA-LPC) and 2-arachidonoyl-lysophosphatidylethanolamine (2-AA-LPE). Next, we identified these two eicosanoid-lysophospholipids in murine myocardium and in isolated platelets. Moreover, we observed robust increases in 2-AA-LPC, 2-AA-LPE, and their downstream 12-LOX oxidation products, 12(S)-HETE-LPC and 12(S)-HETE-LPE, in calcium ionophore (A23187)-stimulated murine platelets. Mechanistically, genetic ablation of iPLA2γ markedly decreased the calcium-stimulated production of 2-AA-LPC, 2-AA-LPE, and 12-HETE-lysophospholipids in mouse platelets. Importantly, a potent and selective 12-LOX inhibitor, ML355, significantly inhibited the production of 12-HETE-LPC and 12-HETE-LPE in activated platelets. Furthermore, we found that aging is accompanied by significant changes in 12-HETE-LPC in murine serum that were also markedly attenuated by iPLA2γ genetic ablation. Collectively, these results identify previously unknown iPLA2γ-initiated signaling pathways mediated by direct 12-LOX oxidation of 2-AA-LPC and 2-AA-LPE. This oxidation generates previously unrecognized eicosanoid-lysophospholipids that may serve as biomarkers for age-related diseases and could potentially be used as targets in therapeutic interventions.
Collapse
Affiliation(s)
- Xinping Liu
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Harold F Sims
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Shaoping Guan
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Beverly G Dilthey
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Richard W Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110; Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri 63110; Department of Chemistry, Washington University, Saint Louis, Missouri 63130.
| |
Collapse
|
20
|
Hajeyah AA, Griffiths WJ, Wang Y, Finch AJ, O’Donnell VB. The Biosynthesis of Enzymatically Oxidized Lipids. Front Endocrinol (Lausanne) 2020; 11:591819. [PMID: 33329396 PMCID: PMC7711093 DOI: 10.3389/fendo.2020.591819] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022] Open
Abstract
Enzymatically oxidized lipids are a specific group of biomolecules that function as key signaling mediators and hormones, regulating various cellular and physiological processes from metabolism and cell death to inflammation and the immune response. They are broadly categorized as either polyunsaturated fatty acid (PUFA) containing (free acid oxygenated PUFA "oxylipins", endocannabinoids, oxidized phospholipids) or cholesterol derivatives (oxysterols, steroid hormones, and bile acids). Their biosynthesis is accomplished by families of enzymes that include lipoxygenases (LOX), cyclooxygenases (COX), cytochrome P450s (CYP), and aldo-keto reductases (AKR). In contrast, non-enzymatically oxidized lipids are produced by uncontrolled oxidation and are broadly considered to be harmful. Here, we provide an overview of the biochemistry and enzymology of LOXs, COXs, CYPs, and AKRs in humans. Next, we present biosynthetic pathways for oxylipins, oxidized phospholipids, oxysterols, bile acids and steroid hormones. Last, we address gaps in knowledge and suggest directions for future work.
Collapse
Affiliation(s)
- Ali A. Hajeyah
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
- *Correspondence: Ali A. Hajeyah,
| | - William J. Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Andrew J. Finch
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Valerie B. O’Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
21
|
Colombo S, Criscuolo A, Zeller M, Fedorova M, Domingues MR, Domingues P. Analysis of oxidised and glycated aminophospholipids: Complete structural characterisation by C30 liquid chromatography-high resolution tandem mass spectrometry. Free Radic Biol Med 2019; 144:144-155. [PMID: 31150763 DOI: 10.1016/j.freeradbiomed.2019.05.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
The aminophospholipids (APL), phosphatidylethanolamine (PE) and phosphatidylserine (PS) are widely present in cell membranes and lipoproteins. Glucose and reactive oxygen species (ROS), such as the hydroxyl radical (•OH), can react with APL leading to an array of oxidised, glycated and glycoxidised derivatives. Modified APL have been implicated in inflammatory diseases and diabetes, and were identified as signalling molecules regulating cell death. However, the biological relevance of these molecules has not been completely established, since they are present in very low amounts, and new sensitive methodologies are needed to detect them in biological systems. Few studies have focused on the characterisation of APL modifications using liquid chromatography-tandem mass spectrometry (LC-MS/MS), mainly using C5 or C18 reversed phase (RP) columns. In the present study, we propose a new analytical approach for the characterisation of complex mixtures of oxidised, glycated and glycoxidised PE and PS. This LC approach was based on a reversed-phase C30 column combined with high-resolution MS, and higher energy C-trap dissociation (HCD) MS/MS. C30 RP-LC separated short and long fatty acyl oxidation products, along with glycoxidised APL bearing oxidative modifications on the glucose moiety and the fatty acyl chains. Functional isomers (e.g. hydroxy-hydroperoxy-APL and tri-hydroxy-APL) and positional isomers (e.g. 9-hydroxy-APL and 13-hydroxy-APL) were also discriminated by the method. HCD fragmentation patterns allowed unequivocal structural characterisation of the modified APL, and are translatable into targeted MS/MS fingerprinting of the modified derivatives in biological samples.
Collapse
Affiliation(s)
- Simone Colombo
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Angela Criscuolo
- Thermo Fisher Scientific, Hanna-Kunath-Straße 11, 28199, Bremen, Germany; Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Germany; Center for Biotechnology and Biomedicine, Universität Leipzig, Germany
| | - Martin Zeller
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Germany; Center for Biotechnology and Biomedicine, Universität Leipzig, Germany
| | - M Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal; Department of Chemistry & CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
22
|
Colombo S, Domingues P, Domingues MR. Mass spectrometry strategies to unveil modified aminophospholipids of biological interest. MASS SPECTROMETRY REVIEWS 2019; 38:323-355. [PMID: 30597614 DOI: 10.1002/mas.21584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 06/09/2023]
Abstract
The biological functions of modified aminophospholipids (APL) have become a topic of interest during the last two decades, and distinct roles have been found for these biomolecules in both physiological and pathological contexts. Modifications of APL include oxidation, glycation, and adduction to electrophilic aldehydes, altogether contributing to a high structural variability of modified APL. An outstanding technique used in this challenging field is mass spectrometry (MS). MS has been widely used to unveil modified APL of biological interest, mainly when associated with soft ionization methods (electrospray and matrix-assisted laser desorption ionization) and coupled with separation techniques as liquid chromatography. This review summarizes the biological roles and the chemical mechanisms underlying APL modifications, and comprehensively reviews the current MS-based knowledge that has been gathered until now for their analysis. The interpretation of the MS data obtained by in vitro-identification studies is explained in detail. The perspective of an analytical detection of modified APL in clinical samples is explored, highlighting the fundamental role of MS in unveiling APL modifications and their relevance in pathophysiology.
Collapse
Affiliation(s)
- Simone Colombo
- Mass Spectrometry Centre, Department of Chemistry and QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry and QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry and QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
- Department of Chemistry and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
23
|
Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer 2019; 19:405-414. [PMID: 31101865 DOI: 10.1038/s41568-019-0149-1] [Citation(s) in RCA: 819] [Impact Index Per Article: 136.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ferroptosis is a recently recognized cell death modality that is morphologically, biochemically and genetically distinct from other forms of cell death and that has emerged to play an important role in cancer biology. Recent discoveries have highlighted the metabolic plasticity of cancer cells and have provided intriguing insights into how metabolic rewiring is a critical event for the persistence, dedifferentiation and expansion of cancer cells. In some cases, this metabolic reprogramming has been linked to an acquired sensitivity to ferroptosis, thus opening up new opportunities to treat therapy-insensitive tumours. However, it is not yet clear what metabolic determinants are critical for therapeutic resistance and evasion of immune surveillance. Therefore, a better understanding of the processes that regulate ferroptosis sensitivity should ultimately aid in the discovery of novel therapeutic strategies to improve cancer treatment. In this Perspectives article, we provide an overview of the known mechanisms that regulate sensitivity to ferroptosis in cancer cells and how the modulation of metabolic pathways controlling ferroptosis might reshape the tumour niche, leading to an immunosuppressive microenvironment that promotes tumour growth and progression.
Collapse
Affiliation(s)
| | - Dmitri V Krysko
- Department of Human Structure and Repair, Ghent University and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
24
|
Liu GY, Moon SH, Jenkins CM, Sims HF, Guan S, Gross RW. Synthesis of oxidized phospholipids by sn-1 acyltransferase using 2-15-HETE lysophospholipids. J Biol Chem 2019; 294:10146-10159. [PMID: 31080170 DOI: 10.1074/jbc.ra119.008766] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/10/2019] [Indexed: 01/13/2023] Open
Abstract
Recently, oxidized phospholipid species have emerged as important signaling lipids in activated immune cells and platelets. The canonical pathway for the synthesis of oxidized phospholipids is through the release of arachidonic acid by cytosolic phospholipase A2α (cPLA2α) followed by its enzymatic oxidation, activation of the carboxylate anion by acyl-CoA synthetase(s), and re-esterification to the sn-2 position by sn-2 acyltransferase activity (i.e. the Lands cycle). However, recent studies have demonstrated the unanticipated significance of sn-1 hydrolysis of arachidonoyl-containing choline and ethanolamine glycerophospholipids by other phospholipases to generate the corresponding 2-arachidonoyl-lysolipids. Herein, we identified a pathway for oxidized phospholipid synthesis comprising sequential sn-1 hydrolysis by a phospholipase A1 (e.g. by patatin-like phospholipase domain-containing 8 (PNPLA8)), direct enzymatic oxidation of the resultant 2-arachidonoyl-lysophospholipids, and the esterification of oxidized 2-arachidonoyl-lysophospholipids by acyl-CoA-dependent sn-1 acyltransferase(s). To circumvent ambiguities associated with acyl migration or hydrolysis, we developed a synthesis for optically active (d- and l-enantiomers) nonhydrolyzable analogs of 2-arachidonoyl-lysophosphatidylcholine (2-AA-LPC). sn-1 acyltransferase activity in murine liver microsomes stereospecifically and preferentially utilized the naturally occurring l-enantiomer of the ether analog of lysophosphatidylcholine. Next, we demonstrated the high selectivity of the sn-1 acyltransferase activity for saturated acyl-CoA species. Importantly, we established that 2-15-hydroxyeicosatetraenoic acid (HETE) ether-LPC sn-1 esterification is markedly activated by thrombin treatment of murine platelets to generate oxidized PC. Collectively, these findings demonstrate the enantiomeric specificity and saturated acyl-CoA selectivity of microsomal sn-1 acyltransferase(s) and reveal its participation in a previously uncharacterized pathway for the synthesis of oxidized phospholipids with cell-signaling properties.
Collapse
Affiliation(s)
- Gao-Yuan Liu
- From the Department of Chemistry, Washington University, Saint Louis, Missouri 63130 and.,Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine
| | - Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine
| | | | - Harold F Sims
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine
| | - Shaoping Guan
- Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine
| | - Richard W Gross
- From the Department of Chemistry, Washington University, Saint Louis, Missouri 63130 and .,Division of Bioorganic Chemistry and Molecular Pharmacology, Department of Medicine.,Developmental Biology, and.,Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110
| |
Collapse
|
25
|
Phospholipid membranes drive abdominal aortic aneurysm development through stimulating coagulation factor activity. Proc Natl Acad Sci U S A 2019; 116:8038-8047. [PMID: 30944221 PMCID: PMC6475397 DOI: 10.1073/pnas.1814409116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a disease of the abdominal aorta where inflammation causes damage and can ultimately lead to rupture. When this happens, uncontrolled internal bleeding can lead to death within minutes. Many aneurysms are not detected until they rupture, and for those that are, treatments to stop them progressing are limited. Here we used biophysics and genetically modified mice to show that a new family of lipids (fats) made by circulating blood cells promote AAA formation in the vessel wall because they directly regulate blood clotting. An approach that prevents AAA development was identified, based on intravenous administration of lipids. The studies provide insights into how AAA develops and may lead to novel therapies for this disease. Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease with high mortality and limited treatment options. How blood lipids regulate AAA development is unknown. Here lipidomics and genetic models demonstrate a central role for procoagulant enzymatically oxidized phospholipids (eoxPL) in regulating AAA. Specifically, through activating coagulation, eoxPL either promoted or inhibited AAA depending on tissue localization. Ang II administration to ApoE−/− mice increased intravascular coagulation during AAA development. Lipidomics revealed large numbers of eoxPL formed within mouse and human AAA lesions. Deletion of eoxPL-generating enzymes (Alox12 or Alox15) or administration of the factor Xa inhibitor rivaroxaban significantly reduced AAA. Alox-deficient mice displayed constitutively dysregulated hemostasis, including a consumptive coagulopathy, characterized by compensatory increase in prothrombotic aminophospholipids (aPL) in circulating cell membranes. Intravenously administered procoagulant PL caused clotting factor activation and depletion, induced a bleeding defect, and significantly reduced AAA development. These data suggest that Alox deletion reduces AAA through diverting coagulation away from the vessel wall due to eoxPL deficiency, instead activating clotting factor consumption and depletion in the circulation. In mouse whole blood, ∼44 eoxPL molecular species formed within minutes of clot initiation. These were significantly elevated with ApoE−/− deletion, and many were absent in Alox−/− mice, identifying specific eoxPL that modulate AAA. Correlation networks demonstrated eoxPL belonged to subfamilies defined by oxylipin composition. Thus, procoagulant PL regulate AAA development through complex interactions with clotting factors. Modulation of the delicate balance between bleeding and thrombosis within either the vessel wall or circulation was revealed that can either drive or prevent disease development.
Collapse
|
26
|
O'Donnell VB, Aldrovandi M, Murphy RC, Krönke G. Enzymatically oxidized phospholipids assume center stage as essential regulators of innate immunity and cell death. Sci Signal 2019; 12:12/574/eaau2293. [PMID: 30914483 DOI: 10.1126/scisignal.aau2293] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enzymatically oxidized phospholipids (eoxPLs) are formed through regulated processes by which eicosanoids or prostaglandins are attached to phospholipids (PLs) in immune cells. These eoxPLs comprise structurally diverse families of biomolecules with potent bioactivities, and they have important immunoregulatory roles in both health and disease. The formation of oxPLs through enzymatic pathways and their signaling capabilities are emerging concepts. This paradigm is changing our understanding of eicosanoid, prostaglandin, and PL biology in health and disease. eoxPLs have roles in cellular events such as ferroptosis, apoptosis, and blood clotting and diseases such as arthritis, diabetes, and cardiovascular disease. They are increasingly recognized as endogenous bioactive mediators and potential targets for drug development. This review will describe recent evidence that places eoxPLs and their biosynthetic pathways center stage in immunoregulation.
Collapse
Affiliation(s)
- Valerie B O'Donnell
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK.
| | - Maceler Aldrovandi
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU) 91054, Erlangen, Germany
| |
Collapse
|
27
|
Lipid Metabolites: The Alarm Signal to Trigger Liver Ischemia-reperfusion Injury. Transplantation 2019; 102:887-889. [PMID: 29782474 DOI: 10.1097/tp.0000000000002206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
28
|
Singh NK, Rao GN. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog Lipid Res 2019; 73:28-45. [PMID: 30472260 PMCID: PMC6338518 DOI: 10.1016/j.plipres.2018.11.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
12/15-lipoxygenase (12/15-LOX) is an enzyme, which oxidizes polyunsaturated fatty acids, particularly omega-6 and -3 fatty acids, to generate a number of bioactive lipid metabolites. A large number of studies have revealed the importance of 12/15-LOX role in oxidative and inflammatory responses. The in vitro studies have demonstrated the ability of 12/15-LOX metabolites in the expression of various genes and production of cytokine related to inflammation and resolution of inflammation. The studies with the use of knockout and transgenic animals for 12/15-LOX have further shown its involvement in the pathogenesis of a variety of human diseases, including cardiovascular, renal, neurological and metabolic disorders. This review summarizes our current knowledge on the role of 12/15-LOX in inflammation and various human diseases.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA.
| |
Collapse
|
29
|
Modulation of the inflammatory response of immune cells in human peripheral blood by oxidized arachidonoyl aminophospholipids. Arch Biochem Biophys 2018; 660:64-71. [PMID: 30315768 DOI: 10.1016/j.abb.2018.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/18/2018] [Accepted: 10/09/2018] [Indexed: 01/06/2023]
Abstract
Aminophospholipids (APL), phosphatidylethanolamine (PE) and phosphatidylserine (PS), can be oxidized upon oxidative stress. Oxidized PE and PS have been detected in clinical samples of different pathologies and may act as modulators of the inflammatory response. However, few studies have focused on the effects of oxidized APL (ox-APL) esterified with arachidonic acid, even though a considerable number of studies have assessed the modulation of the immune system by oxidized 1-palmitoyl-2-arachidonoyl-sn-3-glycerophosphocholine (OxPAPC). In the present study, we have used flow cytometry to evaluate the ability of oxidized PAPE (OxPAPE) and PAPS (OxPAPS) to promote or suppress an inflammatory phenotype on monocytes subsets and myeloid dendritic cells (mDCs). The results indicate that OxPAPE increases the frequency of all monocyte subpopulations expressing TNF-α, which promotes an inflammatory response. However, immune cell stimulation with OxPAPE in the presence of LPS results in a decrease of TNF-α expressed by classical monocytes. Incubation with OxPAPS and LPS induces a decrease in TNF-α produced by monocytes, and a significant decrease in IL-1β expressed by monocytes and mDCs, indicating that OxPAPS reduces the LPS-induced pro-inflammatory expression in these populations. These results show the importance of OxPAPE and OxPAPS as modulators of the inflammatory response and demonstrate their possible contribution to the onset and resolution of human diseases related to oxidative stress and inflammation.
Collapse
|
30
|
Maiorino M, Conrad M, Ursini F. GPx4, Lipid Peroxidation, and Cell Death: Discoveries, Rediscoveries, and Open Issues. Antioxid Redox Signal 2018; 29:61-74. [PMID: 28462584 DOI: 10.1089/ars.2017.7115] [Citation(s) in RCA: 441] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Iron-dependent lipid peroxidation is a complex oxidative process where phospholipid hydroperoxides (PLOOH) are produced in membranes and finally transformed into a series of decomposition products, some of which are endowed with biological activity. It is specifically prevented by glutathione peroxidase 4 (GPx4), the selenoenzyme that reduces PLOOH by glutathione (GSH). PLOOH is both a product and the major initiator of peroxidative chain reactions, as well as an activator of lipoxygenases. α-Tocopherol both specifically breaks peroxidative chain propagation and inhibits lipoxygenases. Thus, GPx4, GSH, and α-tocopherol are integrated in a concerted anti-peroxidant mechanism. Recent Advances: Ferroptosis has been recently identified as a cell death subroutine that is specifically activated by missing GPx4 activity and inhibited by iron chelation or α-tocopherol supplementation. Ferroptosis induction may underlie spontaneous human diseases, such as major neurodegeneration and neuroinflammation, causing an excessive cell death. The basic mechanism of ferroptosis, therefore, fits the features of activation of lipid peroxidation. CRITICAL ISSUES Still lacking are convincing proofs that lipoxygenases are involved in ferroptosis. Also, unknown are the molecules eventually killing cells and the mechanisms underlying the drop of the cellular anti-peroxidant capacity. FUTURE DIRECTIONS Molecular events and mechanisms of ferroptosis to be unraveled and validated on animal models are GPx4 inactivation, role of GSH concentration, increased iron availability, and membrane structure and composition. This is expected to drive drug discovery that is aimed at halting cell death in degenerative diseases or boosting it in cancer cells. Antioxid. Redox Signal. 29, 61-74.
Collapse
Affiliation(s)
- Matilde Maiorino
- 1 Department of Molecular Medicine, University of Padova , Padova, Italy
| | - Marcus Conrad
- 2 Institute of Developmental Genetics , Helmholtz Zentrum München, Neuherberg, Germany
| | - Fulvio Ursini
- 1 Department of Molecular Medicine, University of Padova , Padova, Italy
| |
Collapse
|
31
|
O'Donnell VB, Rossjohn J, Wakelam MJ. Phospholipid signaling in innate immune cells. J Clin Invest 2018; 128:2670-2679. [PMID: 29683435 DOI: 10.1172/jci97944] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Phospholipids comprise a large body of lipids that define cells and organelles by forming membrane structures. Importantly, their complex metabolism represents a highly controlled cellular signaling network that is essential for mounting an effective innate immune response. Phospholipids in innate cells are subject to dynamic regulation by enzymes, whose activities are highly responsive to activation status. Along with their metabolic products, they regulate multiple aspects of innate immune cell biology, including shape change, aggregation, blood clotting, and degranulation. Phospholipid hydrolysis provides substrates for cell-cell communication, enables regulation of hemostasis, immunity, thrombosis, and vascular inflammation, and is centrally important in cardiovascular disease and associated comorbidities. Phospholipids themselves are also recognized by innate-like T cells, which are considered essential for recognition of infection or cancer, as well as self-antigens. This Review describes the major phospholipid metabolic pathways present in innate immune cells and summarizes the formation and metabolism of phospholipids as well as their emerging roles in cell biology and disease.
Collapse
Affiliation(s)
- Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jamie Rossjohn
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, and.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
32
|
Slatter DA, Percy CL, Allen-Redpath K, Gajsiewicz JM, Brooks NJ, Clayton A, Tyrrell VJ, Rosas M, Lauder SN, Watson A, Dul M, Garcia-Diaz Y, Aldrovandi M, Heurich M, Hall J, Morrissey JH, Lacroix-Desmazes S, Delignat S, Jenkins PV, Collins PW, O'Donnell VB. Enzymatically oxidized phospholipids restore thrombin generation in coagulation factor deficiencies. JCI Insight 2018; 3:98459. [PMID: 29563336 PMCID: PMC5926910 DOI: 10.1172/jci.insight.98459] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/16/2018] [Indexed: 12/11/2022] Open
Abstract
Hemostatic defects are treated using coagulation factors; however, clot formation also requires a procoagulant phospholipid (PL) surface. Here, we show that innate immune cell–derived enzymatically oxidized phospholipids (eoxPL) termed hydroxyeicosatetraenoic acid–phospholipids (HETE-PLs) restore hemostasis in human and murine conditions of pathological bleeding. HETE-PLs abolished blood loss in murine hemophilia A and enhanced coagulation in factor VIII- (FVIII-), FIX-, and FX-deficient human plasma . HETE-PLs were decreased in platelets from patients after cardiopulmonary bypass (CPB). To explore molecular mechanisms, the ability of eoxPL to stimulate individual isolated coagulation factor/cofactor complexes was tested in vitro. Extrinsic tenase (FVIIa/tissue factor [TF]), intrinsic tenase (FVIIIa/FIXa), and prothrombinase (FVa/FXa) all were enhanced by both HETE-PEs and HETE-PCs, suggesting a common mechanism involving the fatty acid moiety. In plasma, 9-, 15-, and 12-HETE-PLs were more effective than 5-, 11-, or 8-HETE-PLs, indicating positional isomer specificity. Coagulation was enhanced at lower lipid/factor ratios, consistent with a more concentrated area for protein binding. Surface plasmon resonance confirmed binding of FII and FX to HETE-PEs. HETE-PEs increased membrane curvature and thickness, but not surface charge or homogeneity, possibly suggesting increased accessibility to cations/factors. In summary, innate immune-derived eoxPL enhance calcium-dependent coagulation factor function, and their potential utility in bleeding disorders is proposed. Innate immune-derived enzymatically oxidized phospholipids enhance calcium-dependent coagulation factor function.
Collapse
Affiliation(s)
- David A Slatter
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Charles L Percy
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Keith Allen-Redpath
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Joshua M Gajsiewicz
- Departments of Biological Chemistry and Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nick J Brooks
- Faculty of Natural Science, Department of Chemistry, Imperial College London, London, United Kingdom
| | - Aled Clayton
- Institute of Cancer and Genetics, Velindre Cancer Centre, School of Medicine, and
| | - Victoria J Tyrrell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Marcela Rosas
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Sarah N Lauder
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Andrew Watson
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Maria Dul
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - Yoel Garcia-Diaz
- School of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Maceler Aldrovandi
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Meike Heurich
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Judith Hall
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - James H Morrissey
- Departments of Biological Chemistry and Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - P Vincent Jenkins
- Haematology Department, University Hospital of Wales, Cardiff, United Kingdom
| | - Peter W Collins
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
33
|
Colombo S, Coliva G, Kraj A, Chervet JP, Fedorova M, Domingues P, Domingues MR. Electrochemical oxidation of phosphatidylethanolamines studied by mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2018; 53:223-233. [PMID: 29282829 DOI: 10.1002/jms.4056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/29/2017] [Accepted: 12/18/2017] [Indexed: 06/07/2023]
Affiliation(s)
- Simone Colombo
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Giulia Coliva
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, Leipzig, Germany
| | | | | | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Universität Leipzig, Leipzig, Germany
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
34
|
Lauder SN, Allen-Redpath K, Slatter DA, Aldrovandi M, O'Connor A, Farewell D, Percy CL, Molhoek JE, Rannikko S, Tyrrell VJ, Ferla S, Milne GL, Poole AW, Thomas CP, Obaji S, Taylor PR, Jones SA, de Groot PG, Urbanus RT, Hörkkö S, Uderhardt S, Ackermann J, Vince Jenkins P, Brancale A, Krönke G, Collins PW, O'Donnell VB. Networks of enzymatically oxidized membrane lipids support calcium-dependent coagulation factor binding to maintain hemostasis. Sci Signal 2017; 10:10/507/eaan2787. [PMID: 29184033 DOI: 10.1126/scisignal.aan2787] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Blood coagulation functions as part of the innate immune system by preventing bacterial invasion, and it is critical to stopping blood loss (hemostasis). Coagulation involves the external membrane surface of activated platelets and leukocytes. Using lipidomic, genetic, biochemical, and mathematical modeling approaches, we found that enzymatically oxidized phospholipids (eoxPLs) generated by the activity of leukocyte or platelet lipoxygenases (LOXs) were required for normal hemostasis and promoted coagulation factor activities in a Ca2+- and phosphatidylserine (PS)-dependent manner. In wild-type mice, hydroxyeicosatetraenoic acid-phospholipids (HETE-PLs) enhanced coagulation and restored normal hemostasis in clotting-deficient animals genetically lacking p12-LOX or 12/15-LOX activity. Murine platelets generated 22 eoxPL species, all of which were missing in the absence of p12-LOX. Humans with the thrombotic disorder antiphospholipid syndrome (APS) had statistically significantly increased HETE-PLs in platelets and leukocytes, as well as greater HETE-PL immunoreactivity, than healthy controls. HETE-PLs enhanced membrane binding of the serum protein β2GP1 (β2-glycoprotein 1), an event considered central to the autoimmune reactivity responsible for APS symptoms. Correlation network analysis of 47 platelet eoxPL species in platelets from APS and control subjects identified their enzymatic origin and revealed a complex network of regulation, with the abundance of 31 p12-LOX-derived eoxPL molecules substantially increased in APS. In summary, circulating blood cells generate networks of eoxPL molecules, including HETE-PLs, which change membrane properties to enhance blood coagulation and contribute to the excessive clotting and immunoreactivity of patients with APS.
Collapse
Affiliation(s)
- Sarah N Lauder
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Keith Allen-Redpath
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - David A Slatter
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Maceler Aldrovandi
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Anne O'Connor
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Daniel Farewell
- Division of Population Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Charles L Percy
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Jessica E Molhoek
- Department of Clinical Chemistry and Haematology, University of Utrecht, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Sirpa Rannikko
- Department of Medical Microbiology and Immunology, Research Unit of Biomedicine, Finland and Medical Research Center, University of Oulu, P.O. Box 5000, Oulu 90220, Finland.,Nordlab Oulu, University Hospital, Oulu 90220, Finland
| | - Victoria J Tyrrell
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Salvatore Ferla
- Welsh School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF14 4XN, UK
| | - Ginger L Milne
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Alastair W Poole
- School of Physiology, Pharmacy and Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Christopher P Thomas
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK.,Welsh School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF14 4XN, UK
| | - Samya Obaji
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Philip R Taylor
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Simon A Jones
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Phillip G de Groot
- Department of Clinical Chemistry and Haematology, University of Utrecht, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Rolf T Urbanus
- Department of Clinical Chemistry and Haematology, University of Utrecht, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Sohvi Hörkkö
- Department of Medical Microbiology and Immunology, Research Unit of Biomedicine, Finland and Medical Research Center, University of Oulu, P.O. Box 5000, Oulu 90220, Finland.,Nordlab Oulu, University Hospital, Oulu 90220, Finland
| | - Stefan Uderhardt
- Department of Internal Medicine and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Jochen Ackermann
- Department of Internal Medicine and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany
| | - P Vince Jenkins
- Institute of Molecular Medicine, St James's Hospital, Dublin, Ireland
| | - Andrea Brancale
- Welsh School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF14 4XN, UK
| | - Gerhard Krönke
- Department of Internal Medicine and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Peter W Collins
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. .,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. .,Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
35
|
Wenzel SE, Tyurina YY, Zhao J, St Croix CM, Dar HH, Mao G, Tyurin VA, Anthonymuthu TS, Kapralov AA, Amoscato AA, Mikulska-Ruminska K, Shrivastava IH, Kenny EM, Yang Q, Rosenbaum JC, Sparvero LJ, Emlet DR, Wen X, Minami Y, Qu F, Watkins SC, Holman TR, VanDemark AP, Kellum JA, Bahar I, Bayır H, Kagan VE. PEBP1 Wardens Ferroptosis by Enabling Lipoxygenase Generation of Lipid Death Signals. Cell 2017; 171:628-641.e26. [PMID: 29053969 PMCID: PMC5683852 DOI: 10.1016/j.cell.2017.09.044] [Citation(s) in RCA: 653] [Impact Index Per Article: 81.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/05/2017] [Accepted: 09/25/2017] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a form of programmed cell death that is pathogenic to several acute and chronic diseases and executed via oxygenation of polyunsaturated phosphatidylethanolamines (PE) by 15-lipoxygenases (15-LO) that normally use free polyunsaturated fatty acids as substrates. Mechanisms of the altered 15-LO substrate specificity are enigmatic. We sought a common ferroptosis regulator for 15LO. We discovered that PEBP1, a scaffold protein inhibitor of protein kinase cascades, complexes with two 15LO isoforms, 15LO1 and 15LO2, and changes their substrate competence to generate hydroperoxy-PE. Inadequate reduction of hydroperoxy-PE due to insufficiency or dysfunction of a selenoperoxidase, GPX4, leads to ferroptosis. We demonstrated the importance of PEBP1-dependent regulatory mechanisms of ferroptotic death in airway epithelial cells in asthma, kidney epithelial cells in renal failure, and cortical and hippocampal neurons in brain trauma. As master regulators of ferroptotic cell death with profound implications for human disease, PEBP1/15LO complexes represent a new target for drug discovery.
Collapse
Affiliation(s)
- Sally E Wenzel
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jinming Zhao
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Haider H Dar
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gaowei Mao
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tamil S Anthonymuthu
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexandr A Kapralov
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew A Amoscato
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karolina Mikulska-Ruminska
- Department of Computational and System Biology, University of Pittsburgh, Pittsburgh, PA, USA; Institute of Physics, Nicolaus Copernicus University, Torun, Poland
| | - Indira H Shrivastava
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Computational and System Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elizabeth M Kenny
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qin Yang
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joel C Rosenbaum
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Louis J Sparvero
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - David R Emlet
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyan Wen
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yoshinori Minami
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Feng Qu
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Theodore R Holman
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA, USA
| | - Andrew P VanDemark
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- Department of Computational and System Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayır
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Critical Care Medicine, Center for Critical Care Nephrology, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
36
|
Aoyagi R, Ikeda K, Isobe Y, Arita M. Comprehensive analyses of oxidized phospholipids using a measured MS/MS spectra library. J Lipid Res 2017; 58:2229-2237. [PMID: 28874441 DOI: 10.1194/jlr.d077123] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/26/2017] [Indexed: 11/20/2022] Open
Abstract
Oxidized phospholipids (OxPLs) are widely held to be associated with various diseases, such as arteriosclerosis, diabetes, and cancer. To characterize the structure-specific behavior of OxPLs and their physiological relevance, we developed a comprehensive analytical method by establishing a measured MS/MS spectra library of OxPLs. Biogenic OxPLs were prepared by the addition of specific oxidized fatty acids to cultured cells, where they were incorporated into cellular phospholipids, and untargeted lipidomics by LC-quadrupole/TOF-MS was applied to collect MS/MS spectra for the OxPLs. Based on the measured MS/MS spectra for about 400 molecular species of the biogenic OxPLs, we developed a broad-targeted lipidomics system using triple quadrupole MS. Separation precision of structural isomers was optimized by multiple reaction monitoring analysis and this system enabled us to detect OxPLs at levels as low as 10 fmol. When applied to biological samples, i.e., mouse peritoneal macrophages, this system enabled us to monitor a series of OxPLs endogenously produced in a 12/15-lipoxygenase-dependent manner. This advanced analytical method will be useful to elucidate the structure-specific behavior of OxPLs and their physiological relevance in vivo.
Collapse
Affiliation(s)
- Ryohei Aoyagi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Yosuke Isobe
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa 230-0045, Japan .,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-0011, Japan
| |
Collapse
|
37
|
O'Donnell VB, Murphy RC. Directing eicosanoid esterification into phospholipids. J Lipid Res 2017; 58:837-839. [PMID: 28242788 DOI: 10.1194/jlr.c075986] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CV14 4XN, UK
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO
| |
Collapse
|
38
|
DioxolaneA3-phosphatidylethanolamines are generated by human platelets and stimulate neutrophil integrin expression. Redox Biol 2017; 11:663-672. [PMID: 28160743 PMCID: PMC5288459 DOI: 10.1016/j.redox.2017.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 11/23/2022] Open
Abstract
Activated platelets generate an eicosanoid proposed to be 8-hydroxy-9,10-dioxolane A3 (DXA3). Herein, we demonstrate that significant amounts of DXA3 are rapidly attached to phosphatidylethanolamine (PE) forming four esterified eicosanoids, 16:0p, 18:0p, 18:1p and 18:0a/DXA3-PEs that can activate neutrophil integrin expression. These lipids comprise the majority of DXA3 generated by platelets, are formed in ng amounts (24.3±6.1ng/2×108) and remain membrane bound. Pharmacological studies revealed DXA3-PE formation involves cyclooxygenase-1 (COX), protease-activated receptors (PAR) 1 and 4, cytosolic phospholipase A2 (cPLA2), phospholipase C and intracellular calcium. They are generated primarily via esterification of newly formed DXA3, but can also be formed in vitro via co-oxidation of PE during COX-1 co-oxidation of arachidonate. All four DXA3-PEs were detected in human clots. Purified platelet DXA3-PE activated neutrophil Mac-1 expression, independently of its hydrolysis to the free eicosanoid. This study demonstrates the structures and cellular synthetic pathway for a family of leukocyte-activating platelet phospholipids generated on acute activation, adding to the growing evidence that enzymatic PE oxidation is a physiological event in innate immune cells.
Collapse
|
39
|
The impact of impaired macrophage functions in cystic fibrosis disease progression. J Cyst Fibros 2016; 16:443-453. [PMID: 27856165 DOI: 10.1016/j.jcf.2016.10.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 01/29/2023]
Abstract
The underlying cause of morbidity in cystic fibrosis (CF) is the decline in lung function, which results in part from chronic inflammation. Inflammation and infection occur early in infancy in CF and the role of innate immune defense in CF has been highlighted in the last years. Once thought simply to be consumers of bacteria, macrophages have emerged as highly sensitive immune cells that are located at the balance point between inflammation and resolution of this inflammation in CF pathophysiology. In order to assess the potential role of macrophage in CF, we review the evidence that: (1) CF macrophage has a dysregulated inflammatory phenotype; (2) CF macrophage presents altered phagocytosis capacity and bacterial killing; and (3) lipid disorders in CF macrophage affect its function. These alterations of macrophage weaken innate defense of CF patients and may be involved in CF disease progression and lung damage.
Collapse
|
40
|
Ackermann JA, Hofheinz K, Zaiss MM, Krönke G. The double-edged role of 12/15-lipoxygenase during inflammation and immunity. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:371-381. [PMID: 27480217 DOI: 10.1016/j.bbalip.2016.07.014] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/01/2016] [Accepted: 07/28/2016] [Indexed: 01/18/2023]
Abstract
12/15-Lipoxygenase (12/15-LOX) mediates the enzymatic oxidation of polyunsaturated fatty acids, thereby contributing to the generation of various bioactive lipid mediators. Although 12/15-LOX has been implicated in the pathogenesis of multiple chronic inflammatory diseases, its physiologic functions seem to include potent immune modulatory properties that physiologically contribute to the resolution of inflammation and the clearance of inflammation-associated tissue damage. This review aims to give a comprehensive overview about our current knowledge on the role of this enzyme during the regulation of inflammation and immunity. This article is part of a Special Issue entitled: Lipid modification and lipid peroxidation products in innate immunity and inflammation edited by Christoph J. Binder.
Collapse
Affiliation(s)
- Jochen A Ackermann
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Katharina Hofheinz
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
41
|
Ryman V, Pighetti G, Lippolis J, Gandy J, Applegate C, Sordillo L. Quantification of bovine oxylipids during intramammary Streptococcus uberis infection. Prostaglandins Other Lipid Mediat 2015; 121:207-17. [DOI: 10.1016/j.prostaglandins.2015.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/09/2015] [Accepted: 09/24/2015] [Indexed: 12/19/2022]
|
42
|
Manček-Keber M, Frank-Bertoncelj M, Hafner-Bratkovič I, Smole A, Zorko M, Pirher N, Hayer S, Kralj-Iglič V, Rozman B, Ilc N, Horvat S, Jerala R. Toll-like receptor 4 senses oxidative stress mediated by the oxidation of phospholipids in extracellular vesicles. Sci Signal 2015; 8:ra60. [PMID: 26082436 DOI: 10.1126/scisignal.2005860] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oxidative stress produced in response to infection or sterile injury activates the innate immune response. We found that extracellular vesicles (EVs) isolated from the plasma of patients with rheumatoid arthritis or secreted from cells subjected to oxidative stress contained oxidized phospholipids that stimulated cells expressing Toll-like receptor 4 (TLR4) in a manner dependent on its co-receptor MD-2. EVs from healthy subjects or reconstituted synthetic EVs subjected to limited oxidation gained the ability to stimulate TLR4-expressing cells, whereas prolonged oxidation abrogated this property. Furthermore, we found that 15-lipoxygenase generated hydro(pero)xylated phospholipids that stimulated TLR4-expressing cells. Molecular modeling suggested that the mechanism of activation of TLR4 by oxidized phospholipids in EVs was structurally similar to that of the TLR4 ligand lipopolysaccharide (LPS). This was supported by experiments showing that EV-mediated stimulation of cells required MD-2, that mutations that block LPS binding to TLR4 abrogated the stimulatory effect of EVs, and that EVs induced TLR4 dimerization. On the other hand, analysis of gene expression profiles showed that genes encoding factors that resolve inflammation were more abundantly expressed in responses to EVs than in response to LPS. Together, these data suggest that EVs act as an oxidative stress-induced endogenous danger signal that underlies the pervasive role of TLR4 in inflammatory diseases.
Collapse
Affiliation(s)
- Mateja Manček-Keber
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia. Excellent NMR Future Innovation for Sustainable Technologies, Centre of Excellence, 1000 Ljubljana, Slovenia.
| | - Mojca Frank-Bertoncelj
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia. Excellent NMR Future Innovation for Sustainable Technologies, Centre of Excellence, 1000 Ljubljana, Slovenia
| | - Anže Smole
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Mateja Zorko
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Nina Pirher
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Silvia Hayer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Veronika Kralj-Iglič
- Laboratoryof Clinical Biophysics, Faculty of Health Sciences, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Blaž Rozman
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Nejc Ilc
- Faculty of Computer and Information Science, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia. Department of Animal Science, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Biotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia. Excellent NMR Future Innovation for Sustainable Technologies, Centre of Excellence, 1000 Ljubljana, Slovenia.
| |
Collapse
|
43
|
Rothe T, Gruber F, Uderhardt S, Ipseiz N, Rössner S, Oskolkova O, Blüml S, Leitinger N, Bicker W, Bochkov VN, Yamamoto M, Steinkasserer A, Schett G, Zinser E, Krönke G. 12/15-Lipoxygenase-mediated enzymatic lipid oxidation regulates DC maturation and function. J Clin Invest 2015; 125:1944-54. [PMID: 25844901 DOI: 10.1172/jci78490] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/27/2015] [Indexed: 11/17/2022] Open
Abstract
DCs are able to undergo rapid maturation, which subsequently allows them to initiate and orchestrate T cell-driven immune responses. DC maturation must be tightly controlled in order to avoid random T cell activation and development of autoimmunity. Here, we determined that 12/15-lipoxygenase-meditated (12/15-LO-mediated) enzymatic lipid oxidation regulates DC activation and fine-tunes consecutive T cell responses. Specifically, 12/15-LO activity determined the DC activation threshold via generation of phospholipid oxidation products that induced an antioxidative response dependent on the transcription factor NRF2. Deletion of the 12/15-LO-encoding gene or pharmacologic inhibition of 12/15-LO in murine or human DCs accelerated maturation and shifted the cytokine profile, thereby favoring the differentiation of Th17 cells. Exposure of 12/15-LO-deficient DCs to 12/15-LO-derived oxidized phospholipids attenuated both DC activation and the development of Th17 cells. Analysis of lymphatic tissues from 12/15-LO-deficient mice confirmed enhanced maturation of DCs as well as an increased differentiation of Th17 cells. Moreover, experimental autoimmune encephalomyelitis in mice lacking 12/15-LO resulted in an exacerbated Th17-driven autoimmune disease. Together, our data reveal that 12/15-LO controls maturation of DCs and implicate enzymatic lipid oxidation in shaping the adaptive immune response.
Collapse
|
44
|
Abstract
The inflammatory response plays an important role in host defense and maintenance of homeostasis, while imbalances in these responses can also lead to pathologic disease processes. Emerging data show that RKIP interacts with multiple signaling molecules that may potentiate multiple functions during inflammatory processes. Here, we review the interaction of RKIP with both the MAPK and NF-κB pathways in relation to chronic inflammatory diseases. In these settings, it can both inhibit inflammatory pathways as well contribute to pro-inflammatory signaling, often depending on the interactions with multiple proteins and perhaps lipids. The interactions of RKIP with proteins, phospholipids, fatty acids, and their enzymes thus could play a substantial role in diseases like asthma and diabetes. Targeting interactions of RKIP with these pathways could lead to novel approaches to treatment.
Collapse
Affiliation(s)
- Jinming Zhao
- University of Pittsburgh Asthma Institute at UPMC/Pulmonary Allergy and Critical Care Medicine Division, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sally Wenzel
- University of Pittsburgh Asthma Institute at UPMC/Pulmonary Allergy and Critical Care Medicine Division, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
45
|
Kato S, Nakagawa K, Suzuki Y, Asai A, Nagao M, Nagashima K, Oikawa S, Miyazawa T. Liquid chromatography–tandem mass spectrometry determination of human plasma 1-palmitoyl-2-hydroperoxyoctadecadienoyl-phosphatidylcholine isomers via promotion of sodium adduct formation. Anal Biochem 2015; 471:51-60. [DOI: 10.1016/j.ab.2014.10.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 01/06/2023]
|
46
|
Morgan AH, Hammond VJ, Sakoh-Nakatogawa M, Ohsumi Y, Thomas CP, Blanchet F, Piguet V, Kiselyov K, O'Donnell VB. A novel role for 12/15-lipoxygenase in regulating autophagy. Redox Biol 2014; 4:40-7. [PMID: 25498966 PMCID: PMC4309860 DOI: 10.1016/j.redox.2014.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 11/28/2022] Open
Abstract
12/15-Lipoxygenase (LOX) enzymatically generates oxidized phospholipids in monocytes and macrophages. Herein, we show that cells deficient in 12/15-LOX contain defective mitochondria and numerous cytoplasmic vacuoles containing electron dense material, indicating defects in autophagy or membrane processing, However, both LC3 expression and lipidation were normal both basally and on chloroquine treatment. A LOX-derived oxidized phospholipid, 12-hydroxyeicosatetraenoic acid-phosphatidylethanolamine (12-HETE-PE) was found to be a preferred substrate for yeast Atg8 lipidation, versus native PE, while both native and oxidized PE were effective substrates for LC3 lipidation. Last, phospholipidomics demonstrated altered levels of several phospholipid classes. Thus, we show that oxidized phospholipids generated by 12/15-LOX can act as substrates for key proteins required for effective autophagy and that cells deficient in this enzyme show evidence of autophagic dysfunction. The data functionally link phospholipid oxidation with autophagy for the first time. 12/15-Lipoxygenase-deficient macrophages show evidence of autophagic dysfunction. 12-HETE-PE is a substrate for LC2 and Atg8 lipidation. Macrophages deficient in 12/15-lipoxygenase show altered phospholipid content.
Collapse
Affiliation(s)
- Alwena H Morgan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Victoria J Hammond
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | | | - Yoshinori Ohsumi
- Frontier Research Center, Tokyo Institute of Technology, Nagatsuta 4259-S2-12, Yokohama, Japan
| | - Christopher P Thomas
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Fabien Blanchet
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Vincent Piguet
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Langley Hall, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Valerie B O'Donnell
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| |
Collapse
|
47
|
Davies SS, Guo L. Lipid peroxidation generates biologically active phospholipids including oxidatively N-modified phospholipids. Chem Phys Lipids 2014; 181:1-33. [PMID: 24704586 DOI: 10.1016/j.chemphyslip.2014.03.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/28/2014] [Accepted: 03/18/2014] [Indexed: 12/25/2022]
Abstract
Peroxidation of membranes and lipoproteins converts "inert" phospholipids into a plethora of oxidatively modified phospholipids (oxPL) that can act as signaling molecules. In this review, we will discuss four major classes of oxPL: mildly oxygenated phospholipids, phospholipids with oxidatively truncated acyl chains, phospholipids with cyclized acyl chains, and phospholipids that have been oxidatively N-modified on their headgroups by reactive lipid species. For each class of oxPL we will review the chemical mechanisms of their formation, the evidence for their formation in biological samples, the biological activities and signaling pathways associated with them, and the catabolic pathways for their elimination. We will end by briefly highlighting some of the critical questions that remain about the role of oxPL in physiology and disease.
Collapse
Affiliation(s)
- Sean S Davies
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States.
| | - Lilu Guo
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University, United States
| |
Collapse
|
48
|
O'Donnell VB, Murphy RC, Watson SP. Platelet lipidomics: modern day perspective on lipid discovery and characterization in platelets. Circ Res 2014; 114:1185-203. [PMID: 24677238 PMCID: PMC4021279 DOI: 10.1161/circresaha.114.301597] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lipids are diverse families of biomolecules that perform essential structural and signaling roles in platelets. Their formation and metabolism are tightly controlled by enzymes and signal transduction pathways, and their dysregulation leads to significant defects in platelet function and disease. Platelet activation is associated with significant changes to membrane lipids, and formation of diverse bioactive lipids plays essential roles in hemostasis. In recent years, new generation mass spectrometry analysis of lipids (termed lipidomics) has begun to alter our understanding of how these molecules participate in key cellular processes. Although the application of lipidomics to platelet biology is still in its infancy, seminal earlier studies have shaped our knowledge of how lipids regulate key aspects of platelet biology, including aggregation, shape change, coagulation, and degranulation, as well as how lipids generated by platelets influence other cells, such as leukocytes and the vascular wall, and thus how they regulate hemostasis, vascular integrity, and inflammation, as well as contribute to pathologies, including arterial/deep vein thrombosis and atherosclerosis. This review will provide a brief historical perspective on the characterization of lipids in platelets, then an overview of the new generation lipidomic approaches, their recent application to platelet biology, and future perspectives for research in this area. The major platelet-regulatory lipid families, their formation, metabolism, and their role in health and disease, will be summarized.
Collapse
Affiliation(s)
- Valerie B O'Donnell
- From the Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom (V.B.O'D.); Department of Pharmacology, University of Colorado at Denver, Aurora (R.C.M.); and Birmingham Platelet Group, Centre for Cardiovascular Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, Birmingham, United Kingdom (S.P.W.)
| | | | | |
Collapse
|
49
|
Gonzalez-Juarrero M, Mima N, Trunck LA, Schweizer HP, Bowen RA, Dascher K, Mwangi W, Eckstein TM. Polar lipids of Burkholderia pseudomallei induce different host immune responses. PLoS One 2013; 8:e80368. [PMID: 24260378 PMCID: PMC3832426 DOI: 10.1371/journal.pone.0080368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/08/2013] [Indexed: 12/12/2022] Open
Abstract
Melioidosis is a disease in tropical and subtropical regions of the world that is caused by Burkholderia pseudomallei. In endemic regions the disease occurs primarily in humans and goats. In the present study, we used the goat as a model to dissect the polar lipids of B. pseudomallei to identify lipid molecules that could be used for adjuvants/vaccines or as diagnostic tools. We showed that the lipidome of B. pseudomallei and its fractions contain several polar lipids with the capacity to elicit different immune responses in goats, namely rhamnolipids and ornithine lipids which induced IFN-γ, whereas phospholipids and an undefined polar lipid induced strong IL-10 secretion in CD4+ T cells. Autologous T cells co-cultured with caprine dendritic cells (cDCs) and polar lipids of B. pseudomallei proliferated and up-regulated the expression of CD25 (IL-2 receptor) molecules. Furthermore, we demonstrated that polar lipids were able to up-regulate CD1w2 antigen expression in cDCs derived from peripheral blood monocytes. Interestingly, the same polar lipids had only little effect on the expression of MHC class II DR antigens in the same caprine dendritic cells. Finally, antibody blocking of the CD1w2 molecules on cDCs resulted in decreased expression for IFN-γ by CD4+ T cells. Altogether, these results showed that polar lipids of B. pseudomallei are recognized by the caprine immune system and that their recognition is primarily mediated by the CD1 antigen cluster.
Collapse
Affiliation(s)
- Mercedes Gonzalez-Juarrero
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
| | - Naoko Mima
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
| | - Lily A. Trunck
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
| | - Herbert P. Schweizer
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kyle Dascher
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
| | - Waithaka Mwangi
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Torsten M. Eckstein
- Department of Microbiology Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United State of America
- * E-mail:
| |
Collapse
|
50
|
Abstract
Oxidized PLs (OxPLs) generated in health and disease are now recognized as important mediators of cellular signalling. There is an increasing body of evidence showing that PL peroxidation is not only increased in vascular disorders, but is also a physiological event of relevance to coagulation, innate immunity, and self-tolerance. Nonenzymatically formed OxPLs generated during chronic inflammation is an uncontrolled event, generating hundreds of diverse structures, and prone to more deleterious bioactivities. In contrast, enzymatic formation of OxPLs is tightly regulated, involving receptors and intracellular signaling, acting as part of the normal physiological response to injury in order to restore homeostasis. In the present review, the major nonenzymatic OxPLs structures found during vascular inflammation are summarized, along with a brief description of their known biological activities. Also, we review what is currently known about enzymatic formation of OxPLs by acutely activated immune cells and their signaling actions under homeostatic and pathological conditions.
Collapse
Affiliation(s)
- Maceler Aldrovandi
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| | | |
Collapse
|