1
|
Gou Y, Lv BH, Zhang JF, Li SM, Hei XP, Liu JJ, Li L, Yang JZ, Feng K. Identifying early predictive and diagnostic biomarkers and exploring metabolic pathways for sepsis after trauma based on an untargeted metabolomics approach. Sci Rep 2025; 15:12068. [PMID: 40199964 PMCID: PMC11978901 DOI: 10.1038/s41598-025-92631-3] [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: 11/04/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Systemic inflammatory response syndrome (SIRS) and organ dysfunction make it challenging to predict which major trauma patients are at risk of developing sepsis. Additionally, the unclear pathogenesis of sepsis after trauma contributes to its high morbidity and mortality. Identifying early predictive and diagnostic biomarkers, as well as exploring related metabolic pathways, is crucial for improving early prevention, diagnosis, and treatment. This study prospectively analyzed plasma samples from patients with severe trauma collected between March 2022 and November 2023. Trauma patients were divided into two groups based on whether they developed sepsis within two weeks: the TDDS group (trauma patients who did not develop sepsis) and the TDS group (trauma patients who did develop sepsis). Plasma samples from the TDS group were collected at the time of sepsis diagnosis (Sepsis group). Metabolite concentrations were measured using ultrahigh-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) through untargeted metabolomics. From the differential metabolites between the TDS and TDDS groups, we identified five significant metabolites (all area under the curve (AUC) ≥ 0.94) as early predictive biomarkers for sepsis after trauma: (1) docosatrienoic acid, (2) 7-alpha-carboxy-17-alpha-carboxyethylandrostan lactone phenyl ester, (3) sphingomyelin (SM) 8:1;2O/26:1, (4) N1-[1-(3-isopropenylphenyl)-1-methylethyl]-3-oxobutanamide, and (5) SM 34:2;2O. Furthermore, five significant metabolites (all AUC ≥ 0.85) were identified as early diagnostic biomarkers from the comparison between the TDS and TDDS groups: (1) lysophosphatidylcholine (LPC) O-22:1, (2) LPC O-22:0, (3) uric acid, (4) LPC O-24:2, and (5) LPC 22:0-SN1. 26 metabolites shared between two comparisons (TDS vs. TDDS and sepsis vs. TDS) were identified. Of which, 19 metabolites belong to lipid metabolism. The top three metabolic pathways related to sepsis after trauma under the impact of severe trauma were: (1) glycerophospholipid metabolism, (2) porphyrin metabolism, and (3) sphingolipid metabolism. The top three metabolic pathways related to sepsis after trauma under the impact of infection were: (1) caffeine metabolism, (2) biosynthesis of unsaturated fatty acids, and (3) steroid hormone biosynthesis. Our study identified early predictive and diagnostic biomarkers and explored metabolic pathways related to sepsis after trauma. These findings provide a foundation for future research on the onset and development of sepsis, facilitating its early prevention, diagnosis, and treatment based on specific metabolites and metabolic pathways.
Collapse
Affiliation(s)
- Yi Gou
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Bo-Hui Lv
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China
| | - Sheng-Ming Li
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China
| | - Xiao-Ping Hei
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China
| | - Jing-Jing Liu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Lei Li
- School of Nursing, Guizhou Medical University, Guiyang, 550025, China
| | - Jian-Zhong Yang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| | - Ke Feng
- Department of Emergency Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750003, Ningxia, China.
| |
Collapse
|
2
|
Bao Y, Osowiecka M, Ott C, Tziraki V, Meusburger L, Blaßnig C, Krivda D, Pjevac P, Séneca J, Strauss M, Steffen C, Heck V, Aygün S, Duszka K, Doppelmayer K, Grune T, Pignitter M. Dietary oxidized lipids in redox biology: Oxidized olive oil disrupts lipid metabolism and induces intestinal and hepatic inflammation in C57BL/6J mice. Redox Biol 2025; 81:103575. [PMID: 40043451 PMCID: PMC11927754 DOI: 10.1016/j.redox.2025.103575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Olive oil, rich in oleic acid, is often regarded as a healthier alternative to animal fats high in saturated fatty acids and plant oils rich in oxidizable polyunsaturated fatty acids. However, the redox biological implications and health effects of oxidized olive oil (ox-OO) remain underexplored. Our study investigated its impact on lipid metabolism, intestinal and hepatic inflammation, and gut microbiota. Female C57BL/6J mice were fed either a standard normal (NFD), high-fat diet (HFD), an NFD-ox-OO or HFD-ox-OO, in which ox-OO (180 °C heating, 10 min) was the sole lipid source. Inflammation was assessed using macrophage marker F4/80 immunohistochemical (IHC) staining. Gene expression of inflammatory and lipid metabolism markers (IL-10, NF-kBp65, IL-1β, TNFα, TLR4, COX2, PPARα, PPARγ, CPT1a, SCAD, MCAD, LCAD) was analyzed by qRT-PCR. Soluble epoxide hydrolase (sEH) protein expression was measured using IHC. Oxylipin and carnitine profiles were determined by LC-MS/MS. Gut microbiota was analyzed by 16S rRNA sequencing. Ox-OO disrupted redox homeostasis, leading to lipid metabolic dysfunction in the intestines and liver. In the duodenum and proximal jejunum, ox-OO decreased the levels of anti-inflammatory oxylipins and increased pro-inflammatory mediators, leading to inflammation. In the ileum and colon, ox-OO caused lipid metabolic dysregulation and inflammation. Colon inflammation was linked to inhibited mitochondrial β-oxidation and decreased short-chain fatty acid-producing microbiomes. Notably, redox imbalances were further implicated by the identification of 9,10-epoxy-stearic acid, a novel inflammatory lipid mediator oxidized from dietary oleic acid, which upregulated sEH. Ox-OO affects lipid metabolism and may contribute to inflammation in the gut and liver, raising questions about the assumption that olive oil is always beneficial and suggesting possible risks linked to oxidized oleic acid.
Collapse
Affiliation(s)
- Yifan Bao
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Magdalena Osowiecka
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Vasiliki Tziraki
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Lukas Meusburger
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Claudia Blaßnig
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Daniela Krivda
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Joana Séneca
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
| | - Matthias Strauss
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Doctoral School of Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christina Steffen
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Verena Heck
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Soner Aygün
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Kalina Duszka
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Kevin Doppelmayer
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Tilman Grune
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Marc Pignitter
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Yang C, Wang S, Qi Y, Jin Y, Guan R, Huang Z. Mechanisms of adipocyte regulation: Insights from HADHB gene modulation. PLoS One 2025; 20:e0319384. [PMID: 40146690 PMCID: PMC11949335 DOI: 10.1371/journal.pone.0319384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/31/2025] [Indexed: 03/29/2025] Open
Abstract
The HADHB gene encodes the beta-subunit of 3-hydroxy acyl-CoA dehydrogenase, closely related to energy metabolism, fatty acid synthesis, and catabolism. This study aimed to investigate the effect of the HADHB gene on the proliferation and differentiation of bovine preadipocytes and to gain new insights into the mechanisms of adipocyte regulation. RNA was extracted from adipose tissue of yellow cattle and the HADHB gene CDS region was cloned. Meanwhile, isolation and cultivation of preadipocytes were used for siRNA and adenovirus overexpression, quantitative real-time PCR, transcriptome sequencing, and cell proliferation and cell cycle were measured by oil red staining, CCK8 assay, and flow cytometry. Subsequently, the transcriptome data were analyzed using bioinformatics. The results showed that the HADHB gene modulates significantly the expression of critical genes involved in proliferation (CDK2 and PCNA) and differentiation (PPARγ and CEBPα), influencing preadipocyte proliferation and differentiation and altering cell cycle progression. The results of transcriptome sequencing demonstrated that the overexpression of the HADHB gene markedly altered the transcriptional profile of preadipocytes, with 170 genes exhibiting a significant increase in expression and 113 genes displaying a decrease. The HADHB gene exerts a regulatory influence on the differentiation process of precursor adipocytes by modulating the expression of key genes involved in proliferation and differentiation.These findings highlight the central role of the HADHB gene in adipocyte regulation and provide new insights into the regulatory mechanisms governing adipocyte biology.
Collapse
Affiliation(s)
- Chaoyun Yang
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Shuzhe Wang
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Yunxia Qi
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Yadong Jin
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Ran Guan
- College of Animal Science and Technology, Xichang University, Xichang, China
| | - Zengwen Huang
- College of Animal Science and Technology, Xichang University, Xichang, China
| |
Collapse
|
4
|
Fisk HL, Shaikh SR. Emerging mechanisms of organ crosstalk: The role of oxylipins. NUTR BULL 2025; 50:12-29. [PMID: 39659132 DOI: 10.1111/nbu.12726] [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: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
There is growing interest in the role of oxylipins in the pathophysiology of several diseases. This is accompanied by a limited but evolving evidence base describing augmented oxylipin concentrations in a range of complications including cardiovascular disease, obesity, liver disease and neurological disorders. Despite this, literature describing oxylipin profiles in blood and multiple organs is inconsistent and the mechanisms by which these profiles are altered, and the relationships between localised tissue and circulating oxylipins are poorly understood. Inflammation and immune response associated with disease requires communication across organs and physiological systems. For example, inflammation and comorbidities associated with obesity extend beyond the adipose tissue and affect the vascular, hepatobiliary and digestive systems amongst others. Communication between organs and physiological systems is implicated in the progression of disease as well as the maintenance of homeostasis. There is emerging evidence for the role of oxylipins as a mechanism of communication in organ crosstalk but the role of these in orchestrating multiple organ and system responses is poorly understood. Herein, we review evidence to support and describe the role of oxylipins in organ crosstalk via the cardiosplenic and gut-link axis. In addition, we review emerging mechanisms of oxylipin regulation, the gut microbiome and modification using nutritional intervention. Finally, we describe future perspectives for addressing challenges in measurement and interpretation of oxylipin research with focus on the host genome as a modifier of oxylipin profiles and response to dietary lipid intervention.
Collapse
Affiliation(s)
- Helena Lucy Fisk
- Faculty of Medicine, School of Human Development and Health, Southampton General Hospital, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Zeng Q, Yu Q, Mo Y, Liang H, Chen B, Meng J. Genome-Wide Identification and Functional Characterization of the Acyl-CoA Dehydrogenase (ACAD) Family in Fusarium sacchari. Int J Mol Sci 2025; 26:973. [PMID: 39940743 PMCID: PMC11817166 DOI: 10.3390/ijms26030973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Fusarium sacchari is one of the primary causal agents of Pokkah boeng disease (PBD), an important disease of sugarcane worldwide. The acyl-CoA dehydrogenases (ACADs) constitute a family of flavoenzymes involved in the β-oxidation of fatty acids and amino acid catabolism in mitochondria. However, the role of ACADs in the pathogenesis of F. sacchari is unclear. Here, 14 ACAD-encoding genes (FsACAD-1-FsACAD-14) were identified by screening the entire genome sequence of F. sacchari. The FsACAD genes are distributed across seven chromosomes and were classified into seven clades based on phylogenetic analysis of the protein sequences. In vivo mRNA quantification revealed that the FsACAD genes are differentially expressed during sugarcane infection, and their expression patterns differ significantly in response to the in vitro induction of fatty acids of different classes. Fatty acid utilization assays of the FsACAD-deletion mutants revealed that the FsACADs varied in their preference and ability to break down different fatty acids and amino acids. There was variation in the adverse impact of FsACAD-deletion mutants on fungal traits, including growth, conidiation, stress tolerance, and virulence. These findings provide insights into the roles of FsACADs in F. sacchari, and the identification of FsACADs offers potential new targets for the improved control of PBD.
Collapse
Affiliation(s)
- Quan Zeng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry and Province Co-Sponsored Collaborative Innovation Center for Sugarcane and Sugar Industry, Guangxi University, Nanning 530004, China;
- College of Life Science and Technology, Guangxi University, Nanning 530004, China
| | - Quan Yu
- Guangxi Key Laboratory of Sugarcane Biology, Academy of Sugarcane and Sugar Industry, College of Agriculture, Guangxi University, Nanning 530004, China; (Q.Y.); (Y.M.); (H.L.)
| | - Yingxi Mo
- Guangxi Key Laboratory of Sugarcane Biology, Academy of Sugarcane and Sugar Industry, College of Agriculture, Guangxi University, Nanning 530004, China; (Q.Y.); (Y.M.); (H.L.)
| | - Haoming Liang
- Guangxi Key Laboratory of Sugarcane Biology, Academy of Sugarcane and Sugar Industry, College of Agriculture, Guangxi University, Nanning 530004, China; (Q.Y.); (Y.M.); (H.L.)
| | - Baoshan Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry and Province Co-Sponsored Collaborative Innovation Center for Sugarcane and Sugar Industry, Guangxi University, Nanning 530004, China;
- Guangxi Key Laboratory of Sugarcane Biology, Academy of Sugarcane and Sugar Industry, College of Agriculture, Guangxi University, Nanning 530004, China; (Q.Y.); (Y.M.); (H.L.)
| | - Jiaorong Meng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Ministry and Province Co-Sponsored Collaborative Innovation Center for Sugarcane and Sugar Industry, Guangxi University, Nanning 530004, China;
- Guangxi Key Laboratory of Sugarcane Biology, Academy of Sugarcane and Sugar Industry, College of Agriculture, Guangxi University, Nanning 530004, China; (Q.Y.); (Y.M.); (H.L.)
| |
Collapse
|
6
|
Revol-Cavalier J, Quaranta A, Newman JW, Brash AR, Hamberg M, Wheelock CE. The Octadecanoids: Synthesis and Bioactivity of 18-Carbon Oxygenated Fatty Acids in Mammals, Bacteria, and Fungi. Chem Rev 2025; 125:1-90. [PMID: 39680864 PMCID: PMC11719350 DOI: 10.1021/acs.chemrev.3c00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The octadecanoids are a broad class of lipids consisting of the oxygenated products of 18-carbon fatty acids. Originally referring to production of the phytohormone jasmonic acid, the octadecanoid pathway has been expanded to include products of all 18-carbon fatty acids. Octadecanoids are formed biosynthetically in mammals via cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) activity, as well as nonenzymatically by photo- and autoxidation mechanisms. While octadecanoids are well-known mediators in plants, their role in the regulation of mammalian biological processes has been generally neglected. However, there have been significant advancements in recognizing the importance of these compounds in mammals and their involvement in the mediation of inflammation, nociception, and cell proliferation, as well as in immuno- and tissue modulation, coagulation processes, hormone regulation, and skin barrier formation. More recently, the gut microbiome has been shown to be a significant source of octadecanoid biosynthesis, providing additional biosynthetic routes including hydratase activity (e.g., CLA-HY, FA-HY1, FA-HY2). In this review, we summarize the current field of octadecanoids, propose standardized nomenclature, provide details of octadecanoid preparation and measurement, summarize the phase-I metabolic pathway of octadecanoid formation in mammals, bacteria, and fungi, and describe their biological activity in relation to mammalian pathophysiology as well as their potential use as biomarkers of health and disease.
Collapse
Affiliation(s)
- Johanna Revol-Cavalier
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Alessandro Quaranta
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - John W. Newman
- Western
Human Nutrition Research Center, Agricultural
Research Service, USDA, Davis, California 95616, United States
- Department
of Nutrition, University of California, Davis, Davis, California 95616, United States
- West
Coast Metabolomics Center, Genome Center, University of California, Davis, Davis, California 95616, United States
| | - Alan R. Brash
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mats Hamberg
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Craig E. Wheelock
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm SE-141-86, Sweden
| |
Collapse
|
7
|
Ghorasaini M, Costa D, Tyrrell VJ, Protty M, Giera M, O'Donnell VB. A Method for Analysis of Oxidized Phospholipids from Biological Samples Using Mass Spectrometry. Methods Mol Biol 2025; 2855:155-169. [PMID: 39354307 DOI: 10.1007/978-1-0716-4116-3_10] [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] [Indexed: 10/03/2024]
Abstract
Oxidized phospholipids (oxPLs) are generated during innate immunity and inflammation, where they play a variety of biological roles, including regulation of autoimmunity and coagulation. Some are generated by enzymatic reactions, leading to stereo- and regiospecificity, while many others can be formed through nonenzymatic oxidation and truncation and can be used as biomarkers of oxidative stress. Mass spectrometry methods have been developed over many years for oxPL analysis, which can provide robust estimations of molecular species and amounts, where standards are available. Here we present a method used for the analysis of enzymatically-generated oxPL (eoxPL), which allows quantification of mono-hydroxy oxylipin-containing species. We also show profiling of many other partially characterized structures in tissue samples and provide typical chromatograms obtained.
Collapse
Affiliation(s)
- Mohan Ghorasaini
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniela Costa
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Victoria J Tyrrell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Majd Protty
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Valerie B O'Donnell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
8
|
Protty MB, Tyrrell VJ, Hajeyah AA, Morgan B, Costa D, Li Y, Choudhury A, Mitra R, Bosanquet D, Reed A, Denisenko IK, Nagata K, Shindou H, Cravatt BF, Poole AW, Shimizu T, Yousef Z, Collins PW, O'Donnell VB. Aspirin modulates generation of procoagulant phospholipids in cardiovascular disease, by regulating LPCAT3. J Lipid Res 2025; 66:100727. [PMID: 39674322 PMCID: PMC11754521 DOI: 10.1016/j.jlr.2024.100727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Enzymatically oxygenated phospholipids (eoxPL) from lipoxygenases (LOX) or cyclooxygenase (COX) are prothrombotic. Their generation in arterial disease, and their modulation by cardiovascular therapies is unknown. Furthermore, the Lands cycle acyl-transferases that catalyze their formation are unidentified. eoxPL were measured in platelets and leukocytes from an atherosclerotic cardiovascular disease (ASCVD) cohort and retrieved human arterial thrombi from three anatomical sites. The impact of age, gender, and aspirin was characterized in platelets from healthy subjects administered low-dose aspirin. The role of lysophosphatidylcholine acyltransferase 3 (LPCAT3) in eoxPL biosynthesis was tested using an inhibitor and a cell-free assay. Platelets from ASCVD patients generated lower levels of COX-derived eoxPL but elevated 12-LOX-diacyl forms, than platelets from healthy controls. This associated with aspirin and was recapitulated in healthy subjects by aspirin supplementation. P2Y12 inhibition had no impact on eoxPL. LPCAT3 inhibition selectively prevented 12-LOX-derived diacyl-eoxPL generation. LPCAT3 activity was not directly altered by aspirin. P2Y12 inhibition or aspirin had little impact on eoxPL in leukocytes. Complex aspirin-dependent gender and seasonal effects on platelet eoxPL generation were seen in healthy subjects. Limb or coronary (ST-elevation myocardial infarction, STEMI) thrombi displayed a platelet eoxPL signature while carotid thrombi had a white cell profile. EoxPL are altered in ASCVD by a commonly used cardiovascular therapy, and LPCAT3 was identified as the acyltransferase generating aspirin-sensitive 12-LOX diacyl forms. These changes to the phospholipid composition of blood cells in humans at risk of thrombosis may be clinically significant where the procoagulant membrane plays a central role in driving elevated thrombotic risk.
Collapse
Affiliation(s)
- Majd B Protty
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK.
| | | | - Ali A Hajeyah
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Bethan Morgan
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Daniela Costa
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Yong Li
- Bristol Platelet Group, School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Anirban Choudhury
- Morriston Cardiac Centre, Swansea Bay University Health Board, Swansea, UK
| | - Rito Mitra
- Department of Cardiology, University Hospital of Wales, Cardiff, UK
| | - David Bosanquet
- Department of Vascular Surgery, Aneurin Bevan University Health Board, Cwmbran, UK
| | - Alex Reed
- Department of Chemistry, The Scripps Research Institute, San Diego, CA
| | | | | | - Hideo Shindou
- National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Alastair W Poole
- Bristol Platelet Group, School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Takao Shimizu
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Zaheer Yousef
- Department of Cardiology, University Hospital of Wales, Cardiff, UK
| | - Peter W Collins
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | | |
Collapse
|
9
|
Ağagündüz D, Yeşildemir Ö, Koçyiğit E, Koçak T, Özen Ünaldı B, Ayakdaş G, Budán F. Oxylipins Derived from PUFAs in Cardiometabolic Diseases: Mechanism of Actions and Possible Nutritional Interactions. Nutrients 2024; 16:3812. [PMID: 39599599 PMCID: PMC11597274 DOI: 10.3390/nu16223812] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Oxylipins are oxidized fatty acids, both saturated and unsaturated, formed through pathways that involve singlet oxygen or dioxygen-mediated oxygenation reactions and are primarily produced by enzyme families such as cyclooxygenases, lipoxygenases, and cytochrome P450. These lipid-based complex bioactive molecules are pivotal signal mediators, acting in a hormone-like manner in the pathophysiology of numerous diseases, especially cardiometabolic diseases via modulating plenty of mechanisms. It has been reported that omega-6 and omega-3 oxylipins are important novel biomarkers of cardiometabolic diseases. Moreover, collected literature has noted that diet and dietary components, especially fatty acids, can modulate these oxygenated lipid products since they are mainly derived from dietary omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) or linoleic acid and α-linolenic by elongation and desaturation pathways. This comprehensive review aims to examine their correlations to cardiometabolic diseases and how diets modulate oxylipins. Also, some aspects of developing new biomarkers and therapeutical utilization are detailed in this review.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Türkiye
| | - Özge Yeşildemir
- Department of Nutrition and Dietetics, Bursa Uludag University, Görükle Campus, 16059 Bursa, Türkiye;
| | - Emine Koçyiğit
- Department of Nutrition and Dietetics, Ordu University, Cumhuriyet Yerleşkesi, 52200 Ordu, Türkiye;
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Gümüşhane University, Gümüşhanevî Kampüsü, 29100 Gümüşhane, Türkiye;
| | - Buket Özen Ünaldı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye;
| | - Gamze Ayakdaş
- Department of Nutrition and Dietetics, Acıbadem University, Kerem Aydınlar Campus, 34752 İstanbul, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
10
|
Gao C, Luo L, Fan Y, Guo L, Guo L, Tao L, Tao F, Xu DX, Gibson RA, Makrides M, Wang H, Huang Y. Gestational PFAS exposure and newborn size: The modifying effect of cord blood fatty acids. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2024; 22:100476. [PMID: 39228809 PMCID: PMC11369372 DOI: 10.1016/j.ese.2024.100476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Per- and polyfluoroalkyl substances (PFASs) can disrupt lipid metabolism, and changes in cord blood fatty acid composition have been observed in small newborns. Emerging evidence suggests that exposure to PFASs during pregnancy is linked to decreased newborn size, although the evidence is not consistent. The modifying effect of fatty acids on the associations of gestational PFAS exposure with newborn size is still unknown. Here we show that the nutritional status of the fetus, as indicated by the level of fatty acids in the cord blood, mitigates the adverse effects of gestational PFAS exposure on the size of the newborn. Our study confirms the adverse developmental effects of PFASs and identifies emerging short-chain PFASs as the primary drivers of reduced newborn size, despite their lower exposure burden compared to legacy PFASs. Additionally, we find the protective role of cord blood fatty acids, suggesting potential strategies for mitigating the detrimental effects of emerging environmental exposures on human health. Our findings provide new evidence of the potential toxicity of emerging PFASs and call for further toxicity evaluations of these pollutants for regulatory purposes. Future studies should consider the complex interaction between exposure and nutrition within the human body, particularly during the first thousand days of life, to promote lifelong health.
Collapse
Affiliation(s)
- Chang Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Lin Luo
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Yijun Fan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Liyan Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Lijuan Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Lin Tao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Fangbiao Tao
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
- Department of Maternal & Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230031, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Robert A. Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, 5000, South Australia, Australia
| | - Maria Makrides
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, North Adelaide, 5000, South Australia, Australia
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230031, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230031, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230031, China
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230031, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Clinical Research Center, Suzhou Hospital of Anhui Medical University, Suzhou, 234099, China
| |
Collapse
|
11
|
Ding X, Xu M, Zhang Y, Long C, Su X, Zhang Y, Qiao Y, Zhang X, Zhou Q, Tan G, Ma J. Integration of four-dimensional proteomics and network pharmacology to reveal molecular mechanisms of multi-components multi-targets effects of Sini decoction on myocardial infarction. J Pharm Biomed Anal 2024; 253:116526. [PMID: 39486393 DOI: 10.1016/j.jpba.2024.116526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/17/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024]
Abstract
Sini Decoction (SND) has been proven to be an effective formula to alleviate cardiac injury of myocardial infarction (MI). However, the potential mechanism of SND remains unclear. In this study, the MI rat model was established by ligating the left anterior descending coronary artery. A total of 17 SND-distributed components in heart were identified by using ultra-high performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOFMS). The combination of four-dimensional (4D) proteomics and network pharmacology was employed to find the potential targets for therapeutic intervention, and molecular docking and cellular thermal shift assay (CETSA) were used to reveal the interactions between the potential targets and the potential active components distributed in heart of SND. 33 SND-effected proteins were identified by 4D proteomics, which was involved in carbon metabolism, fatty acid metabolism, valine, leucine and isoleucine degradation, tricarboxylic acid (TCA) cycle and PPAR signaling pathway. 17 potential SND-targeted direct proteins were screened by comparing SND-effected proteins generated from 4D proteomics with the MI-related proteins obtained from disease database. The potential relationships between 17 components and 17 potential SND-targeted direct proteins were established by molecular docking analysis, in which songorine, benzoylhypaconine, hypaconine, formononetin, and liquiritigenin could be bound to the surrounding amino acid residues in the binding pocket of Mtor, Parp1, Acadm, Crat, and Aldh2. Then, CETSA analysis further confirmed that songorine and benzoylhypaconine could increase the heat stability of Mtor and Parp1 in cardiac tissue lysate, respectively, which suggested that there existed direct interactions between songorine and Mtor, and benzoylhypaconine and Parp1. In summary, this work concluded that SND produced cardioprotective effects mainly through preserving energy metabolism, also demonstrated that the combination of 4D proteomics and network pharmacology was a promising tool for uncovering the molecular mechanisms of multi-components multi-targets effects of TCM.
Collapse
Affiliation(s)
- Xin Ding
- Department of traditional Chinese medicine, Xijing Hospital, Air Force Medical University, Xi'an 710032, China; School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Min Xu
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China; The Third Stationed Outpatient Department, General Hospital of Central Theater Command, Wuhan 430070, China
| | - Ya Zhang
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Cuiping Long
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Xuemei Su
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Yang Zhang
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Yan Qiao
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Xingxing Zhang
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Qian Zhou
- Department of traditional Chinese medicine, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| | - Guangguo Tan
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| | - Jing Ma
- Department of traditional Chinese medicine, Xijing Hospital, Air Force Medical University, Xi'an 710032, China.
| |
Collapse
|
12
|
Huff HC, Kim JS, Ojha A, Sinha S, Das A. Real time changes in the expression of eicosanoid synthesizing enzymes during inflammation. Prostaglandins Other Lipid Mediat 2024; 174:106839. [PMID: 38679226 DOI: 10.1016/j.prostaglandins.2024.106839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Immune responses during inflammation involve complex, well-coordinated lipid signaling pathways. Eicosanoids are a class of lipid signaling molecules derived from polyunsaturated fatty acids such as arachidonic acid and constitute a major network that controls inflammation and its subsequent resolution. Arachidonic acid is metabolized by enzymes in three different pathways to form a variety of lipid metabolites that can be either pro- or anti-inflammatory. Therefore, an understanding of the time-dependent gene expression, lipid metabolite profiles and cytokine profiles during the initial inflammatory response is necessary, as it will allow for the design of time-dependent therapeutics. Herein, we investigate the multi-level regulation of this process. After stimulating RAW 264.7 cells, a mouse-derived macrophage cell line commonly used to examine inflammatory responses, we examine the gene expression of 44 relevant lipid metabolizing enzymes from the different eicosanoid synthesizing classes. We also measure the formation of lipid metabolites and production of cytokines at selected time points. Results reveal a dynamic relationship between the time-course of inflammation dependent gene expression of the three eicosanoid synthesizing enzymes.
Collapse
Affiliation(s)
- Hannah C Huff
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA 30332, USA
| | - Justin S Kim
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA 30332, USA
| | - Abhishek Ojha
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Saurabh Sinha
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Aditi Das
- School of Chemistry and Biochemistry, College of Sciences. Georgia Institute of Technology, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, Atlanta, GA 30332, USA.
| |
Collapse
|
13
|
Harlina PW, Maritha V, Yang X, Dixon R, Muchtaridi M, Shahzad R, Nur'Isma EA. Exploring oxylipins in processed foods: Understanding mechanisms, analytical perspectives, and enhancing quality with lipidomics. Heliyon 2024; 10:e35917. [PMID: 39247353 PMCID: PMC11379580 DOI: 10.1016/j.heliyon.2024.e35917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Oxylipins are active lipid compounds formed through the oxidation of unsaturated fatty acids. These compounds have drawn considerable attention due to the potential impact on human health and processed food quality. Therefore, this study aimed to deepen current understanding and assess recent analytical advancements regarding the physiological roles of oxylipins in processed food products using lipidomics. The mechanisms behind oxylipins production in processed foods were extensively investigated, underscoring potential associations with chronic diseases. This indicates the need for innovative strategies to mitigate harmful oxylipins levels to enhance the safety and shelf life of processed food products. The results showed that mitigation methods, including the use of antioxidants and optimization of processing parameters, reduced oxylipins levels. The integration of lipidomics with food safety and quality control processes is evident in cutting-edge methods such as nuclear magnetic resonance and mass spectrometry for compliance and real-time evaluation. Aside from envisioning the future trajectory of food science and industry through prospective studies on oxylipins and processed foods, the results also provide the basis for future investigations, innovation, and advancements in the dynamic field of food science and technology.
Collapse
Affiliation(s)
- Putri Widyanti Harlina
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363, Bandung, Indonesia
| | - Vevi Maritha
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, 45363, Bandung, Indonesia
- Pharmacy Study Program, Faculty of Health and Science, Universitas PGRI Madiun, Indonesia
| | - Xiang Yang
- Department of Animal Science, University of California Davis, California, 95616, United States
| | - Roy Dixon
- Department of Chemistry, California State University, Sacramento, CA, 95819, United States
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, 45363, Bandung, Indonesia
| | - Raheel Shahzad
- Research Center for Genetic Engineering, National Research and Innovation Agency (BRIN), Indonesia
| | - Ernisa Adha Nur'Isma
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363, Bandung, Indonesia
| |
Collapse
|
14
|
Paquay S, Duraffourd J, Bury M, Heremans IP, Caligiore F, Gerin I, Stroobant V, Jacobs J, Pinon A, Graff J, Vertommen D, Van Schaftingen E, Dewulf JP, Bommer GT. ACAD10 and ACAD11 allow entry of 4-hydroxy fatty acids into β-oxidation. Cell Mol Life Sci 2024; 81:367. [PMID: 39174697 PMCID: PMC11342911 DOI: 10.1007/s00018-024-05397-8] [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: 04/27/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Hydroxylated fatty acids are important intermediates in lipid metabolism and signaling. Surprisingly, the metabolism of 4-hydroxy fatty acids remains largely unexplored. We found that both ACAD10 and ACAD11 unite two enzymatic activities to introduce these metabolites into mitochondrial and peroxisomal β-oxidation, respectively. First, they phosphorylate 4-hydroxyacyl-CoAs via a kinase domain, followed by an elimination of the phosphate to form enoyl-CoAs catalyzed by an acyl-CoA dehydrogenase (ACAD) domain. Studies in knockout cell lines revealed that ACAD10 preferentially metabolizes shorter chain 4-hydroxy fatty acids than ACAD11 (i.e. 6 carbons versus 10 carbons). Yet, recombinant proteins showed comparable activity on the corresponding 4-hydroxyacyl-CoAs. This suggests that the localization of ACAD10 and ACAD11 to mitochondria and peroxisomes, respectively, might influence their physiological substrate spectrum. Interestingly, we observed that ACAD10 is cleaved internally during its maturation generating a C-terminal part consisting of the ACAD domain, and an N-terminal part comprising the kinase domain and a haloacid dehalogenase (HAD) domain. HAD domains often exhibit phosphatase activity, but negligible activity was observed in the case of ACAD10. Yet, inactivation of a presumptive key residue in this domain significantly increased the kinase activity, suggesting that this domain might have acquired a regulatory function to prevent accumulation of the phospho-hydroxyacyl-CoA intermediate. Taken together, our work reveals that 4-hydroxy fatty acids enter mitochondrial and peroxisomal fatty acid β-oxidation via two enzymes with an overlapping substrate repertoire.
Collapse
Affiliation(s)
- Stéphanie Paquay
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Pediatric Neurology and Metabolic Diseases, Cliniques Universitaires St. Luc, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Julia Duraffourd
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Marina Bury
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Isaac P Heremans
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Francesco Caligiore
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Isabelle Gerin
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | | | - Jean Jacobs
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Aymeric Pinon
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Julie Graff
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Didier Vertommen
- Protein Phosphorylation Unit, de Duve Institute & MASSPROT Platform, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Emile Van Schaftingen
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Joseph P Dewulf
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Laboratory Medicine, Cliniques Universitaires St. Luc, Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Guido T Bommer
- Metabolic Research Group, de Duve Institute & WELRI, Université Catholique de Louvain, 1200, Brussels, Belgium.
- WELBIO Department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
15
|
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
|
16
|
Parchem K, Letsiou S, Petan T, Oskolkova O, Medina I, Kuda O, O'Donnell VB, Nicolaou A, Fedorova M, Bochkov V, Gladine C. Oxylipin profiling for clinical research: Current status and future perspectives. Prog Lipid Res 2024; 95:101276. [PMID: 38697517 DOI: 10.1016/j.plipres.2024.101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Oxylipins are potent lipid mediators with increasing interest in clinical research. They are usually measured in systemic circulation and can provide a wealth of information regarding key biological processes such as inflammation, vascular tone, or blood coagulation. Although procedures still require harmonization to generate comparable oxylipin datasets, performing comprehensive profiling of circulating oxylipins in large studies is feasible and no longer restricted by technical barriers. However, it is essential to improve and facilitate the biological interpretation of complex oxylipin profiles to truly leverage their potential in clinical research. This requires regular updating of our knowledge about the metabolism and the mode of action of oxylipins, and consideration of all factors that may influence circulating oxylipin profiles independently of the studied disease or condition. This review aims to provide the readers with updated and necessary information regarding oxylipin metabolism, their different forms in systemic circulation, the current limitations in deducing oxylipin cellular effects from in vitro bioactivity studies, the biological and technical confounding factors needed to consider for a proper interpretation of oxylipin profiles.
Collapse
Affiliation(s)
- Karol Parchem
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, 11/12 Gabriela Narutowicza St., 80-233 Gdańsk, Poland; Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 53210 Pardubice, Czech Republic.
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Ag. Spiridonos St. Egaleo, 12243 Athens, Greece.
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia.
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Isabel Medina
- Instituto de Investigaciones Marinas-Consejo Superior de Investigaciones Científicas (IIM-CSIC), Eduardo Cabello 6, E-36208 Vigo, Spain.
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.
| | - Anna Nicolaou
- School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, 01307 Dresden, Germany.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria.
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France.
| |
Collapse
|
17
|
Fan H, Tan Y. Lipid Droplet-Mitochondria Contacts in Health and Disease. Int J Mol Sci 2024; 25:6878. [PMID: 38999988 PMCID: PMC11240910 DOI: 10.3390/ijms25136878] [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: 05/15/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The orchestration of cellular metabolism and redox balance is a complex, multifaceted process crucial for maintaining cellular homeostasis. Lipid droplets (LDs), once considered inert storage depots for neutral lipids, are now recognized as dynamic organelles critical in lipid metabolism and energy regulation. Mitochondria, the powerhouses of the cell, play a central role in energy production, metabolic pathways, and redox signaling. The physical and functional contacts between LDs and mitochondria facilitate a direct transfer of lipids, primarily fatty acids, which are crucial for mitochondrial β-oxidation, thus influencing energy homeostasis and cellular health. This review highlights recent advances in understanding the mechanisms governing LD-mitochondria interactions and their regulation, drawing attention to proteins and pathways that mediate these contacts. We discuss the physiological relevance of these interactions, emphasizing their role in maintaining energy and redox balance within cells, and how these processes are critical in response to metabolic demands and stress conditions. Furthermore, we explore the pathological implications of dysregulated LD-mitochondria interactions, particularly in the context of metabolic diseases such as obesity, diabetes, and non-alcoholic fatty liver disease, and their potential links to cardiovascular and neurodegenerative diseases. Conclusively, this review provides a comprehensive overview of the current understanding of LD-mitochondria interactions, underscoring their significance in cellular metabolism and suggesting future research directions that could unveil novel therapeutic targets for metabolic and degenerative diseases.
Collapse
Affiliation(s)
- Hongjun Fan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yanjie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
18
|
Zeng L, Ma J, Wei T, Wang H, Yang G, Han C, Zhu T, Tian H, Zhang M. The effect of canagliflozin on gut microbiota and metabolites in type 2 diabetic mice. Genes Genomics 2024; 46:541-555. [PMID: 38483772 DOI: 10.1007/s13258-024-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 01/03/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND Sodium glucose cotransporter 2 inhibitor (SGLT2i) represent a new type of hypoglycemic medicine that can cause massive loss of glucose from the urine, which have several benefits of reducing body weight and improving the prognosis of cardiovascular and kidney diseases. Although they are oral medicated hypoglycemic agents, their effects on the gut microbiome and function have been unclear. OBJECTIVE In order to describe the effects of canagliflozin on intestinal flora and metabolites, diabetic mice were randomized to receive canagliflozin or isoconcentration carboxymethylcellulose sodium by gavage for 8 weeks. Feces were collected for 16 S rRNA gene and LC-MS/MS analysis and enriched metabolic pathways through Kyoto Encyclopedia of Genes and Genomes (KEGG). Liver, muscle, intestinal, fat were collected for qRT-PCR according to KEGG enriched metabolic pathways. RESULTS Our results showed that canagliflozin significantly increased GLP-1 level and impacted on the composition of gut microbiota and metabolites. It mainly increased Muribaculum, Ruminococcaceae_UCG_014, Lachnospiraceae-UCG-001, decreased ursodeoxycholic acids (UDCA) and hyodeoxycholic acids (HDCA), and increased fatty acids metabolites in feces. CONCLUSION In conclusion, we analyzed the changes of intestinal microbial composition and metabolites in diabetic mice after canagliflozin intervention and found that canagliflozin influenced intestinal fatty acid and bile acid (BA) metabolism. This study will provide reference for subsequent SGLT2i and intestinal related research.
Collapse
Affiliation(s)
- Li Zeng
- Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jideng Ma
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Tiantian Wei
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hao Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Guitao Yang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Chongxiang Han
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Tao Zhu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Haoming Tian
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Min Zhang
- Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
19
|
Gao R, Yang K, Le S, Chen H, Sun X, Dong Z, Gao P, Wang X, Shi J, Qu Y, Wei X, Hu K, Wang J, Jin L, Li Y, Ge J, Sun A. Aldehyde dehydrogenase 2 serves as a key cardiometabolic adaptation regulator in response to plateau hypoxia in mice. Transl Res 2024; 267:25-38. [PMID: 38181846 DOI: 10.1016/j.trsl.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/08/2023] [Accepted: 12/22/2023] [Indexed: 01/07/2024]
Abstract
High-altitude heart disease (HAHD) is a complex pathophysiological condition related to systemic hypobaric hypoxia in response to transitioning to high altitude. Hypoxia can cause myocardial metabolic dysregulation, leading to an increased risk of heart failure and sudden cardiac death. Aldehyde dehydrogenase 2 (ALDH2) could regulate myocardial energy metabolism and plays a protective role in various cardiovascular diseases. This study aims to determine the effects of plateau hypoxia (PH) on cardiac metabolism and function, investigate the associated role of ALDH2, and explore potential therapeutic targets. We discovered that PH significantly reduced survival rate and cardiac function. These effects were exacerbated by ALDH2 deficiency. PH also caused a shift in the myocardial fuel source from fatty acids to glucose; ALDH2 deficiency impaired this adaptive metabolic shift. Untargeted/targeted metabolomics and transmission electron microscopy revealed that ALDH2 deficiency promoted myocardial fatty-acid deposition, leading to enhanced fatty-acid transport, lipotoxicity and mitochondrial dysfunction. Furthermore, results showed that ALDH2 attenuated PH-induced impairment of adaptive metabolic programs through 4-HNE/CPT1 signaling, and the CPT1 inhibitor etomoxir significantly ameliorated ALDH2 deficiency-induced cardiac impairment and improved survival in PH mice. Together, our data reveal ALDH2 acts as a key cardiometabolic adaptation regulator in response to PH. CPT1 inhibitor, etomoxir, may attenuate ALDH2 deficiency-induced effects and improved cardiac function in response to PH.
Collapse
Affiliation(s)
- Rifeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cardiac Surgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China; Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Kun Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiguan Le
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Hanchuan Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xilu Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanan Qu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Wei
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yi Li
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Hateley C, Olona A, Halliday L, Edin ML, Ko JH, Forlano R, Terra X, Lih FB, Beltrán-Debón R, Manousou P, Purkayastha S, Moorthy K, Thursz MR, Zhang G, Goldin RD, Zeldin DC, Petretto E, Behmoaras J. Multi-tissue profiling of oxylipins reveal a conserved up-regulation of epoxide:diol ratio that associates with white adipose tissue inflammation and liver steatosis in obesity. EBioMedicine 2024; 103:105127. [PMID: 38677183 PMCID: PMC11061246 DOI: 10.1016/j.ebiom.2024.105127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Obesity drives maladaptive changes in the white adipose tissue (WAT) which can progressively cause insulin resistance, type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated liver disease (MASLD). Obesity-mediated loss of WAT homeostasis can trigger liver steatosis through dysregulated lipid pathways such as those related to polyunsaturated fatty acid (PUFA)-derived oxylipins. However, the exact relationship between oxylipins and metabolic syndrome remains elusive and cross-tissue dynamics of oxylipins are ill-defined. METHODS We quantified PUFA-related oxylipin species in the omental WAT, liver biopsies and plasma of 88 patients undergoing bariatric surgery (female N = 79) and 9 patients (female N = 4) undergoing upper gastrointestinal surgery, using UPLC-MS/MS. We integrated oxylipin abundance with WAT phenotypes (adipogenesis, adipocyte hypertrophy, macrophage infiltration, type I and VI collagen remodelling) and the severity of MASLD (steatosis, inflammation, fibrosis) quantified in each biopsy. The integrative analysis was subjected to (i) adjustment for known risk factors and, (ii) control for potential drug-effects through UPLC-MS/MS analysis of metformin-treated fat explants ex vivo. FINDINGS We reveal a generalized down-regulation of cytochrome P450 (CYP)-derived diols during obesity conserved between the WAT and plasma. Notably, epoxide:diol ratio, indicative of soluble epoxide hydrolyse (sEH) activity, increases with WAT inflammation/fibrosis, hepatic steatosis and T2DM. Increased 12,13-EpOME:DiHOME in WAT and liver is a marker of worsening metabolic syndrome in patients with obesity. INTERPRETATION These findings suggest a dampened sEH activity and a possible role of fatty acid diols during metabolic syndrome in major metabolic organs such as WAT and liver. They also have implications in view of the clinical trials based on sEH inhibition for metabolic syndrome. FUNDING Wellcome Trust (PS3431_WMIH); Duke-NUS (Intramural Goh Cardiovascular Research Award (Duke-NUS-GCR/2022/0020); National Medical Research Council (OFLCG22may-0011); National Institute of Environmental Health Sciences (Z01 ES025034); NIHR Imperial Biomedical Research Centre.
Collapse
Affiliation(s)
- Charlotte Hateley
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Antoni Olona
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Laura Halliday
- Department of Surgery and Cancer, Imperial College London, UK
| | - Matthew L Edin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Jeong-Hun Ko
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Ximena Terra
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Fred B Lih
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Raúl Beltrán-Debón
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, MoBioFood Research Group, Tarragona, Spain
| | - Penelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Sanjay Purkayastha
- Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK; University of Brunel, Kingston Lane, Uxbridge, London, UB8 3PH, UK
| | - Krishna Moorthy
- Department of Surgery and Cancer, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Mark R Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Guodong Zhang
- Department of Nutrition, College of Agriculture and Environmental Sciences, 3135 Meyer Hall, One Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Robert D Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, UK; Imperial College Healthcare NHS Trust, St. Mary's Hospital, Praed Street, London, W2 1NY, UK
| | - Darryl C Zeldin
- Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Enrico Petretto
- Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore; Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University (CPU), Nanjing, China
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK; Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
21
|
Gong J. Oxylipins biosynthesis and the regulation of bovine postpartum inflammation. Prostaglandins Other Lipid Mediat 2024; 171:106814. [PMID: 38280540 DOI: 10.1016/j.prostaglandins.2024.106814] [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: 09/16/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Uncontrolled or dysregulated inflammation has adverse effects on the reproduction, production and health of animals, and is a major pathological cause of increased incidence and severity of infectious and metabolic diseases. To achieve successful transition from a non-lactation pregnant state to a non-pregnant lactation state, drastic metabolic and endocrine alteration have taken place in dairy cows during the periparturient period. These physiological changes, coupled with decreased dry matter intake near calving and sudden change of diet composition after calving, have the potential to disrupt the delicate balance between pro- and anti-inflammation, resulting in a disordered or excessive inflammatory response. In addition to cytokines and other immunoregulatory factors, most oxylipins formed from polyunsaturated fatty acids (PUFAs) via enzymatic and nonenzymatic oxygenation pathways have pro- or anti-inflammatory properties and play a pivotal role in the onset, development and resolution of inflammation. However, little attention has been paid to the possibility that oxylipins could function as endogenous immunomodulating agents. This review will provide a detailed overview of the main oxylipins derived from different PUFAs and discuss the regulatory role that oxylipins play in the postpartum inflammatory response in dairy cows. Based on the current research, much remains to be illuminated in this emerging field. Understanding the role that oxylipins play in the control of postpartum inflammation and inflammatory-based disease may improve our ability to prevent transition disorders via Management, pharmacological, genetic selection and dietary intervention strategies.
Collapse
Affiliation(s)
- Jian Gong
- College of Life Science and Technology, Inner Mongolia Normal University, 81 Zhaowuda Road, Hohhot 010022, China.
| |
Collapse
|
22
|
Rashan EH, Bartlett AK, Khana DB, Zhang J, Jain R, Smith AJ, Baker ZN, Cook T, Caldwell A, Chevalier AR, Pfleger BF, Yuan P, Amador-Noguez D, Simcox JA, Pagliarini DJ. ACAD10 and ACAD11 enable mammalian 4-hydroxy acid lipid catabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574893. [PMID: 38260250 PMCID: PMC10802472 DOI: 10.1101/2024.01.09.574893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Fatty acid β-oxidation (FAO) is a central catabolic pathway with broad implications for organismal health. However, various fatty acids are largely incompatible with standard FAO machinery until they are modified by other enzymes. Included among these are the 4-hydroxy acids (4-HAs)-fatty acids hydroxylated at the 4 (γ) position-which can be provided from dietary intake, lipid peroxidation, and certain drugs of abuse. Here, we reveal that two atypical and poorly characterized acyl-CoA dehydrogenases (ACADs), ACAD10 and ACAD11, drive 4-HA catabolism in mice. Unlike other ACADs, ACAD10 and ACAD11 feature kinase domains N-terminal to their ACAD domains that phosphorylate the 4-OH position as a requisite step in the conversion of 4-hydroxyacyl-CoAs into 2-enoyl-CoAs-conventional FAO intermediates. Our ACAD11 cryo-EM structure and molecular modeling reveal a unique binding pocket capable of accommodating this phosphorylated intermediate. We further show that ACAD10 is mitochondrial and necessary for catabolizing shorter-chain 4-HAs, whereas ACAD11 is peroxisomal and enables longer-chain 4-HA catabolism. Mice lacking ACAD11 accumulate 4-HAs in their plasma while comparable 3- and 5-hydroxy acids remain unchanged. Collectively, this work defines ACAD10 and ACAD11 as the primary gatekeepers of mammalian 4-HA catabolism and sets the stage for broader investigations into the ramifications of aberrant 4-HA metabolism in human health and disease.
Collapse
Affiliation(s)
- Edrees H. Rashan
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Abigail K. Bartlett
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Daven B. Khana
- Department of Microbiology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jingying Zhang
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Andrew J. Smith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Zakery N. Baker
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Taylor Cook
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Alana Caldwell
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Autumn R. Chevalier
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Brian F. Pfleger
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Peng Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Daniel Amador-Noguez
- Department of Microbiology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Judith A. Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David J. Pagliarini
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
23
|
Liang N, Harsch BA, Zhou S, Borkowska A, Shearer GC, Kaddurah-Daouk R, Newman JW, Borkowski K. Oxylipin transport by lipoprotein particles and its functional implications for cardiometabolic and neurological disorders. Prog Lipid Res 2024; 93:101265. [PMID: 37979798 DOI: 10.1016/j.plipres.2023.101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Lipoprotein metabolism is critical to inflammation. While the periphery and central nervous system (CNS) have separate yet connected lipoprotein systems, impaired lipoprotein metabolism is implicated in both cardiometabolic and neurological disorders. Despite the substantial investigation into the composition, structure and function of lipoproteins, the lipoprotein oxylipin profiles, their influence on lipoprotein functions, and their potential biological implications are unclear. Lipoproteins carry most of the circulating oxylipins. Importantly, lipoprotein-mediated oxylipin transport allows for endocrine signaling by these lipid mediators, long considered to have only autocrine and paracrine functions. Alterations in plasma lipoprotein oxylipin composition can directly impact inflammatory responses of lipoprotein metabolizing cells. Similar investigations of CNS lipoprotein oxylipins are non-existent to date. However, as APOE4 is associated with Alzheimer's disease-related microglia dysfunction and oxylipin dysregulation, ApoE4-dependent lipoprotein oxylipin modulation in neurological pathologies is suggested. Such investigations are crucial to bridge knowledge gaps linking oxylipin- and lipoprotein-related disorders in both periphery and CNS. Here, after providing a summary of existent literatures on lipoprotein oxylipin analysis methods, we emphasize the importance of lipoproteins in oxylipin transport and argue that understanding the compartmentalization and distribution of lipoprotein oxylipins may fundamentally alter our consideration of the roles of lipoprotein in cardiometabolic and neurological disorders.
Collapse
Affiliation(s)
- Nuanyi Liang
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - Brian A Harsch
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sitong Zhou
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA 95616, USA
| | - Alison Borkowska
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gregory C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke Institute for Brain Sciences and Department of Medicine, Duke University, Durham, NC, 27708, USA; Duke Institute of Brain Sciences, Duke University, Durham, NC, USA; Department of Medicine, Duke University, Durham, NC, USA
| | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA; Department of Nutrition, University of California - Davis, Davis, CA 95616, USA; Western Human Nutrition Research Center, United States Department of Agriculture - Agriculture Research Service, Davis, CA 95616, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
24
|
Pan LA, Naviaux JC, Wang L, Li K, Monk JM, Lingampelly SS, Segreti AM, Bloom K, Vockley J, Tarnopolsky MA, Finegold DN, Peters DG, Naviaux RK. Metabolic features of treatment-refractory major depressive disorder with suicidal ideation. Transl Psychiatry 2023; 13:393. [PMID: 38097555 PMCID: PMC10721812 DOI: 10.1038/s41398-023-02696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
Peripheral blood metabolomics was used to gain chemical insight into the biology of treatment-refractory Major Depressive Disorder with suicidal ideation, and to identify individualized differences for personalized care. The study cohort consisted of 99 patients with treatment-refractory major depressive disorder and suicidal ideation (trMDD-SI n = 52 females and 47 males) and 94 age- and sex-matched healthy controls (n = 48 females and 46 males). The median age was 29 years (IQR 22-42). Targeted, broad-spectrum metabolomics measured 448 metabolites. Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) were measured as biomarkers of mitochondrial dysfunction. The diagnostic accuracy of plasma metabolomics was over 90% (95%CI: 0.80-1.0) by area under the receiver operator characteristic (AUROC) curve analysis. Over 55% of the metabolic impact in males and 75% in females came from abnormalities in lipids. Modified purines and pyrimidines from tRNA, rRNA, and mRNA turnover were increased in the trMDD-SI group. FGF21 was increased in both males and females. Increased lactate, glutamate, and saccharopine, and decreased cystine provided evidence of reductive stress. Seventy-five percent of the metabolomic abnormalities found were individualized. Personalized deficiencies in CoQ10, flavin adenine dinucleotide (FAD), citrulline, lutein, carnitine, or folate were found. Pathways regulated by mitochondrial function dominated the metabolic signature. Peripheral blood metabolomics identified mitochondrial dysfunction and reductive stress as common denominators in suicidal ideation associated with treatment-refractory major depressive disorder. Individualized metabolic differences were found that may help with personalized management.
Collapse
Affiliation(s)
- Lisa A Pan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Panomics Mental Health Initiative, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, 750 Washington Rd, Suite 19, Pittsburgh, PA, 15228, USA.
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Sai Sachin Lingampelly
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Anna Maria Segreti
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kaitlyn Bloom
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - David N Finegold
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- New Hope Molecular, LLC, Pittsburgh, PA, USA
| | - David G Peters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
25
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
Dalli J, Gomez EA. Reply to: Failure to apply standard limit-of-detection or limit-of-quantitation criteria to specialized pro-resolving mediator analysis incorrectly characterizes their presence in biological samples. Nat Commun 2023; 14:7293. [PMID: 37949899 PMCID: PMC10638266 DOI: 10.1038/s41467-023-41767-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 09/18/2023] [Indexed: 11/12/2023] Open
Affiliation(s)
- Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| | - Esteban A Gomez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| |
Collapse
|
27
|
Sun P, Cui M, Jing J, Kong F, Wang S, Tang L, Leng J, Chen K. Deciphering the molecular and cellular atlas of immune cells in septic patients with different bacterial infections. J Transl Med 2023; 21:777. [PMID: 37919720 PMCID: PMC10621118 DOI: 10.1186/s12967-023-04631-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Sepsis is a life-threatening organ dysfunction caused by abnormal immune responses to various, predominantly bacterial, infections. Different bacterial infections lead to substantial variation in disease manifestation and therapeutic strategies. However, the underlying cellular heterogeneity and mechanisms involved remain poorly understood. METHODS Multiple bulk transcriptome datasets from septic patients with 12 types of bacterial infections were integrated to identify signature genes for each infection. Signature genes were mapped onto an integrated large single-cell RNA (scRNA) dataset from septic patients, to identify subsets of cells associated with different sepsis types, and multiple omics datasets were combined to reveal the underlying molecular mechanisms. In addition, an scRNA dataset and spatial transcriptome data were used to identify signaling pathways in sepsis-related cells. Finally, molecular screening, optimization, and de novo design were conducted to identify potential targeted drugs and compounds. RESULTS We elucidated the cellular heterogeneity among septic patients with different bacterial infections. In Escherichia coli (E. coli) sepsis, 19 signature genes involved in epigenetic regulation and metabolism were identified, of which DRAM1 was demonstrated to promote autophagy and glycolysis in response to E. coli infection. DRAM1 upregulation was confirmed in an independent sepsis cohort. Further, we showed that DRAM1 could maintain survival of a pro-inflammatory monocyte subset, C10_ULK1, which induces systemic inflammation by interacting with other cell subsets via resistin and integrin signaling pathways in blood and kidney tissue, respectively. Finally, retapamulin was identified and optimized as a potential drug for treatment of E. coli sepsis targeting the signature gene, DRAM1, and inhibiting E. coli protein synthesis. Several other targeted drugs were also identified in other types of sepsis, including nystatin targeting C1QA in Neisseria sepsis and dalfopristin targeting CTSD in Streptococcus viridans sepsis. CONCLUSION Our study provides a comprehensive overview of the cellular heterogeneity and underlying mechanisms in septic patients with various bacterial infections, providing insights to inform development of stratified targeted therapies for sepsis.
Collapse
Affiliation(s)
- Ping Sun
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Mintian Cui
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Jiongjie Jing
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Fanyu Kong
- Department of Internal Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shixi Wang
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Lunxian Tang
- Department of Internal Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Junling Leng
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Kun Chen
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
28
|
Muniz-Santos R, Lucieri-Costa G, de Almeida MAP, Moraes-de-Souza I, Brito MADSM, Silva AR, Gonçalves-de-Albuquerque CF. Lipid oxidation dysregulation: an emerging player in the pathophysiology of sepsis. Front Immunol 2023; 14:1224335. [PMID: 37600769 PMCID: PMC10435884 DOI: 10.3389/fimmu.2023.1224335] [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: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by abnormal host response to infection. Millions of people are affected annually worldwide. Derangement of the inflammatory response is crucial in sepsis pathogenesis. However, metabolic, coagulation, and thermoregulatory alterations also occur in patients with sepsis. Fatty acid mobilization and oxidation changes may assume the role of a protagonist in sepsis pathogenesis. Lipid oxidation and free fatty acids (FFAs) are potentially valuable markers for sepsis diagnosis and prognosis. Herein, we discuss inflammatory and metabolic dysfunction during sepsis, focusing on fatty acid oxidation (FAO) alterations in the liver and muscle (skeletal and cardiac) and their implications in sepsis development.
Collapse
Affiliation(s)
- Renan Muniz-Santos
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giovanna Lucieri-Costa
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Augusto P. de Almeida
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabelle Moraes-de-Souza
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Czyżowska A, Brown J, Xu H, Sataranatarajan K, Kinter M, Tyrell VJ, O'Donnell VB, Van Remmen H. Elevated phospholipid hydroperoxide glutathione peroxidase (GPX4) expression modulates oxylipin formation and inhibits age-related skeletal muscle atrophy and weakness. Redox Biol 2023; 64:102761. [PMID: 37279604 PMCID: PMC10276143 DOI: 10.1016/j.redox.2023.102761] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
Our previous studies support a key role for mitochondrial lipid hydroperoxides as important contributors to denervation-related muscle atrophy, including muscle atrophy associated with aging. Phospholipid hydroperoxide glutathione peroxidase 4 (GPX4) is an essential antioxidant enzyme that directly reduces phospholipid hydroperoxides and we previously reported that denervation-induced muscle atrophy is blunted in a mouse model of GPX4 overexpression. Therefore, the goal of the present study was to determine whether GPX4 overexpression can reduce the age-related increase in mitochondrial hydroperoxides in skeletal muscle and ameliorate age-related muscle atrophy and weakness (sarcopenia). Male C57Bl6 WT and GPX4 transgenic (GPX4Tg) mice were studied at 3 to 5 and 23-29 months of age. Basal mitochondrial peroxide generation was reduced by 34% in muscle fibers from aged GPX4Tg compared to old WT mice. GPX4 overexpression also reduced levels of lipid peroxidation products: 4-HNE, MDA, and LOOHs by 38%, 32%, and 84% respectively in aged GPX4Tg mice compared to aged WT mice. Muscle mass was preserved in old GPX4 Tg mice by 11% and specific force generation was 21% higher in old GPX4Tg versus age matched male WT mice. Oxylipins from lipoxygenases (LOX) and cyclooxygenase (COX), as well as less abundant non-enzymatically generated isomers, were significantly reduced by GPX4 overexpression. The expression of cPLA2, 12/15-LOX and COX-2 were 1.9-, 10.5- and 3.4-fold greater in old versus young WT muscle respectively, and 12/15-LOX and COX-2 levels were reduced by 37% and 35%, respectively in muscle from old GPX4Tg mice. Our study suggests that lipid peroxidation products may play an important role in the development of sarcopenia, and their detoxification might be an effective intervention in preventing muscle atrophy.
Collapse
Affiliation(s)
- Agnieszka Czyżowska
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jacob Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Kavitha Sataranatarajan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Victoria J Tyrell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, CF14 4XN, UK
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, CF14 4XN, UK
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
30
|
Liao T, Mei W, Zhang L, Ding L, Yang N, Wang P, Zhang L. L-carnitine alleviates synovitis in knee osteoarthritis by regulating lipid accumulation and mitochondrial function through the AMPK-ACC-CPT1 signaling pathway. J Orthop Surg Res 2023; 18:386. [PMID: 37237380 DOI: 10.1186/s13018-023-03872-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a disability-associated condition that is rapidly growing with the increase in obesity rates worldwide. There is a pressing need for precise management and timely intervention in the development of KOA. L-carnitine has been frequently recommended as a supplement to increase physical activity in obese individuals due to its role in fatty acid metabolism, immune disorders, and in maintaining the mitochondrial acetyl-CoA/CoA ratio. In this study, we aimed to investigate the anti-inflammatory effects of L-carnitine on KOA and delineate a potential molecular mechanism. METHODS Lipopolysaccharide-stimulated primary rat fibroblast-like synoviocytes (FLS) were treated with an AMP-activated protein kinase (AMPK) inhibitor or siRNA and carnitine palmitoyltransferase 1 (CPT1) siRNA to examine the synovial protective effects of L-carnitine. An anterior cruciate ligament transection model of rats was treated with an AMPK agonist (metformin) and CPT1 inhibitor (etomoxir) to define the therapeutic effects of L-carnitine. RESULTS L-carnitine displayed a protective effect against synovitis of KOA in vitro and in vivo experiments. Specifically, L-carnitine treatment can reduce synovitis by inhibiting AMPK-ACC-CPT1 pathway activation and showed an increase in fatty acid β-oxidation, a lower lipid accumulation, and a noticeable improvement in mitochondrial function. CONCLUSIONS Our data suggested that L-carnitine can mitigate synovitis in FLS and synovial tissue, and the underlying mechanism may be related to improving mitochondrial function and reducing lipid accumulation via the AMPK-ACC-CPT1 signaling pathway. Therefore, L-carnitine may be a potential treatment strategy for KOA.
Collapse
Affiliation(s)
- Taiyang Liao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Mei
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Li Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liang Ding
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Nan Yang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China.
| | - Li Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
31
|
Polyunsaturated and Saturated Oxylipin Plasma Levels Allow Monitoring the Non-Alcoholic Fatty Liver Disease Progression to Severe Stages. Antioxidants (Basel) 2023; 12:antiox12030711. [PMID: 36978959 PMCID: PMC10045849 DOI: 10.3390/antiox12030711] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Hepatic fat accumulation is the hallmark of non-alcoholic fatty liver disease (NAFLD). Our aim was to determine the plasma levels of oxylipins, free polyunsaturated fatty acids (PUFA) and markers of lipid peroxidation in patients with NAFLD in progressive stages of the pathology. Ninety 40–60-year-old adults diagnosed with metabolic syndrome were distributed in without, mild, moderate or severe NAFLD stages. The free PUFA and oxylipin plasma levels were determined by the UHPLC–MS/MS system. The plasma levels of oxylipins produced by cyclooxygenases, lipoxygenases and cytochrome P450, such as prostaglandin 2α (PGF2α), lipoxinB4 and maresin-1, were higher in severe NAFLD patients, pointing to the coexistence of both inflammation and resolution processes. The plasma levels of the saturated oxylipins 16-hydroxyl-palmitate and 3-hydroxyl-myristate were also higher in the severe NAFLD patients, suggesting a dysregulation of oxidation of fatty acids. The plasma 12-hydroxyl-estearate (12HEST) levels in severe NAFLD were higher than in the other stages, indicating that the hydroxylation of saturated fatty acid produced by reactive oxygen species is more present in this severe stage of NAFLD. The plasma levels of 12HEST and PGF2α are potential candidate biomarkers for diagnosing NAFLD vs. non-NAFLD. In conclusion, the NAFLD progression can be monitored by measuring the plasma levels of free PUFA and oxylipins characterizing the different NAFLD stages or the absence of this disease in metabolic syndrome patients.
Collapse
|
32
|
Aukema HM, Ravandi A. Factors affecting variability in free oxylipins in mammalian tissues. Curr Opin Clin Nutr Metab Care 2023; 26:91-98. [PMID: 36892958 DOI: 10.1097/mco.0000000000000892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF THE REVIEW Along with the growing interest in oxylipins is an increasing awareness of multiple sources of variability in oxylipin data. This review summarizes recent findings that highlight the experimental and biological sources of variation in free oxylipins. RECENT FINDINGS Experimental factors that affect oxylipin variability include different methods of euthanasia, postmortem changes, cell culture reagents, tissue processing conditions and timing, storage losses, freeze-thaw cycles, sample preparation techniques, ion suppression, matrix effects, use and availability of oxylipin standards, and postanalysis procedures. Biological factors include dietary lipids, fasting, supplemental selenium, vitamin A deficiency, dietary antioxidants and the microbiome. Overt, but also more subtle differences in health affect oxylipin levels, including during resolution of inflammation and long-term recovery from disease. Sex, genetic variation, exposure to air pollution and chemicals found in food packaging and household and personal care products, as well as many pharmaceuticals used to treat health conditions also affect oxylipin levels. SUMMARY Experimental sources of oxylipin variability can be minimized with proper analytical procedures and protocol standardization. Fully characterizing study parameters will help delineate biological factors of variability, which are rich sources of information that can be used to probe oxylipin mechanisms of action and to investigate their roles in health.
Collapse
Affiliation(s)
- Harold M Aukema
- Department of Food and Human Nutritional Sciences, University of Manitoba
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Hospital Albrechtsen Research Centre
- Precision Cardiovascular Medicine Group, St Boniface Hospital Albrechtsen Research Centre
| | - Amir Ravandi
- Precision Cardiovascular Medicine Group, St Boniface Hospital Albrechtsen Research Centre
- Department of Physiology and Pathophysiology, University of Manitoba
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
33
|
Qian H, Ge A, Jiang JJ, Xu JF. Necroptosis-related subtypes are associated with bronchiectasis in pulmonary non-tuberculous mycobacteria-infected patients: a perspective based on transcriptomic analysis. Eur J Clin Microbiol Infect Dis 2023; 42:141-152. [PMID: 36469164 DOI: 10.1007/s10096-022-04532-2] [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: 08/24/2022] [Accepted: 11/23/2022] [Indexed: 12/11/2022]
Abstract
The aim of this study was to explore the potential mechanisms responsible for the different manifestations of bronchiectasis in patients with pulmonary non-tuberculous mycobacteria (pNTM) infection. We found that the necroptosis level increased significantly after NTM infection. Further, the 31 pNTM-infected patients were classified into two subtypes based on necroptosis-related genes (NRGs) by unsupervised cluster analysis. After that, we compared the differences in clinical parameters, immune cell infiltration, and gene expression between the two subtypes. We observed that the high-necroptosis subtype possessed higher CT scores for bronchiectasis extent (P = 0.008) and severity (P = 0.023). And, more neutrophil infiltration in the high-necroptosis subtype was demonstrated both by the CIBERSORT algorithm and by blood neutrophil count (P = 0.001). Next, 688 differentially expressed genes (DEGs) between two subtypes were identified. To explore the portion in DEGs that might contribute to bronchiectasis, we intersected the DEGs with two gene modules. These two gene modules were identified as the most associated with CT scores for bronchiectasis extent and severity by weighted gene co-expression network analysis (WGCNA). Ninety-three intersection genes were obtained. Finally, 7 hub genes including ACSL1, ANXA3, DYSF, HK3, SLC11A1, STX11, and TLR4 were further screened out by machine learning algorithms and protein-protein interaction network analysis. These results suggested that the differential levels of necroptosis in pNTM patients might lead to differential extent and severity of bronchiectasis on radiographic imaging. This process might be associated with neutrophil infiltration and the involvement of seven hub genes.
Collapse
Affiliation(s)
- Hao Qian
- Department of Respiratory and Critical Care Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Ai Ge
- Department of Respiratory and Critical Care Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Ji-Jin Jiang
- Department of Respiratory and Critical Care Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China. .,Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
34
|
Fu X, Yin HH, Wu MJ, He X, Jiang Q, Zhang LT, Liu JY. High Sensitivity and Wide Linearity LC-MS/MS Method for Oxylipin Quantification in Multiple Biological Samples. J Lipid Res 2022; 63:100302. [PMID: 36265716 PMCID: PMC9678976 DOI: 10.1016/j.jlr.2022.100302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022] Open
Abstract
Oxylipins are important biological regulators that have received extensive research attention. Due to the extremely low concentrations, large concentration variations, and high structural similarity of many oxylipins, the quantitative analysis of oxylipins in biological samples is always a great challenge. Here, we developed a liquid chromatography-tandem mass spectrometry-based method with high sensitivity, wide linearity, and acceptable resolution for quantitative profiling of oxylipins in multiple biological samples. A total of 104 oxylipins, some with a high risk of detection crosstalk, were well separated on a 150 mm column over 20 min. The method showed high sensitivity with lower limits of quantitation for 87 oxylipins, reaching 0.05-0.5 pg. Unexpectedly, we found that the linear range for 16, 18, and 17 oxylipins reached 10,000, 20,000, and 40,000 folds, respectively. Due to the high sensitivity, while reducing sample consumption to below half the volume of previous methods, 74, 78, and 59 low-abundance oxylipins, among which some were difficult to detect like lipoxins and resolvins, were well quantified in the tested mouse plasma, mouse liver, and human plasma samples, respectively. Additionally, we determined that analytes with multifarious concentrations of over a 1,000-fold difference could be well quantified simultaneously due to the wide linearity. In conclusion, most likely due to the instrumental advancement, this method effectively improves the quantitative sensitivity and linear range over existing methods, which will facilitate and advance the study of the physiological and pathophysiological functions of oxylipins.
Collapse
Affiliation(s)
- Xian Fu
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Hou-Hua Yin
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ming-Jun Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xin He
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Qing Jiang
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ling-Tong Zhang
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jun-Yan Liu
- Center for Novel Target & Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Du J, Cao L, Gao J, Jia R, Zhu H, Nie Z, Xi B, Yin G, Ma Y, Xu G. Protective Effects of Glycyrrhiza Total Flavones on Liver Injury Induced by Streptococcus agalactiae in Tilapia ( Oreochromis niloticus). Antibiotics (Basel) 2022; 11:1648. [PMID: 36421292 PMCID: PMC9686810 DOI: 10.3390/antibiotics11111648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
Clinical studies have confirmed that Glycyrrhiza total flavones (GTFs) have good anti-hepatic injury, but whether they have a good protective effect on anti-hepatic injury activity induced by Streptococcus agalactiae in tilapia (Oreochromis niloticus) is unknown. The aims of this study were to investigate the protective effects of Glycyrrhiza total flavones on liver injury induced by S. agalactiae (SA) and its underlying mechanism in fish. A total of 150 tilapia were randomly divided into five groups, each with three replicates containing 10 fish: normal control group, S. agalactiae infection group, and three Glycyrrhiza total flavone treatment groups (addition of 0.1, 0.5, or 1.0 g of GTF to 1 kg of feed). The normal control group was only fed with basic diet, after 60 d of feeding, and intraperitoneal injection of the same volume of normal saline (0.05 mL/10 g body weight); the S. agalactiae infection group was fed with basic diet, and the S. agalactiae solution was intraperitoneally injected after 60 d of feeding (0.05 mL/10 g body weight); the three GTF treatment groups were fed with a diet containing 0.1, 0.5, or 1.0 g/kg of GTF, and the S. agalactiae solution was intraperitoneally injected after 60 d of feeding (0.05 mL/10 g body weight). After 48 h injection, blood and liver tissues were collected to measure biochemical parameters and mRNA levels to evaluate the liver protection of GTFs. Compared with the control group, the serum levels of glutamic oxaloacetic transaminase (GOT), glutamic pyruvic transaminase (GPT), alkaline phosphatase (AKP) and glucose (GLU) in the streptococcal infection group increased significantly, while the levels of total antioxidant capacity (T-AOC), superoxide dismutase (SOD), catalase (CAT) and reduced glutathione (GSH) decreased significantly; observations of pathological sections showed obvious damage to the liver tissue structure in response to streptococcal infection. S. agalactiae can also cause fatty liver injury, affecting the function of fatty acid β-oxidation and biosynthesis in the liver of tilapia, and also causing damage to function of the immune system. The addition of GTFs to the diet could improve oxidative stress injury caused by S. agalactiae in tilapia liver tissue to different degrees, promote the β-oxidation of fatty acids in the liver, accelerate the lipid metabolism in the liver, and repair the damaged liver tissue. GTFs have a good protective effect on liver injury caused by streptococcus.
Collapse
Affiliation(s)
- Jinliang Du
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
- College of Veterinary Medicine, Agricultural University of Hebei, Baoding 071001, China
| | - Liping Cao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Jiancao Gao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Rui Jia
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Haojun Zhu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Zhijuan Nie
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Bingwen Xi
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Guojun Yin
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Yuzhong Ma
- College of Veterinary Medicine, Agricultural University of Hebei, Baoding 071001, China
| | - Gangchun Xu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| |
Collapse
|
36
|
Tang L, Bai X, Xie X, Chen G, Jia X, Lei M, Li C, Lai S. Negative effects of heat stress on ovarian tissue in female rabbit. Front Vet Sci 2022; 9:1009182. [PMID: 36452142 PMCID: PMC9704112 DOI: 10.3389/fvets.2022.1009182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/27/2022] [Indexed: 07/30/2023] Open
Abstract
Numerous studies have highlighted the role of miRNA in the deformation and necrosis of cells of ovarian tissue caused by heat stress (HS), which ultimately affects ovarian function. Although the role of small RNAs has been investigated in alterations in ovarian tissue functioning in response to HS, the expression profile of ovarian miRNA has been explored to a lesser extent. In this study, female rabbits were subject to HS treatment by using electrical heater. The current work demonstrated that HS could significantly change physiological performance of female rabbits including body weight, rectal temperature and relative ovary weight, and significantly reduce serum IL-2, IL-8, CAT, and GSH-Px concentrations by enzyme-linked immunosorbent assay (ELISA) technique. As a result, an increase in apoptosis in ovarian cells, as well as unhealthy follicles, were observed by Hematoxylin-eosin (HE) and TUNEL staining. Additionally, small RNA-seq revealed changes in the miRNA expression profile of rabbit ovaries under HS. Five hundred fourteen miRNAs were obtained including known miRNAs 442 and novel miRNAs 72. Among these miRNAs, 23 miRNAs were significantly expressed under HS. Eleven differentially expressed miRNAs (DE miRNAs) and 9 their predicted targets were confirmed by qPCR, which were expected miRNA-mRNA negative regulation pattern. Among the DE miRNAs and targets, miR-141-39 may target COQ6, miR-449a-5p and miR-34c-5p may control RFC5 and RTN2 together, miR-449a-5p may target ACADVL, miR-34c-5p potentially targets Bcl-2 and miR-196b-5p potentially regulates CASK and HOXB6. Thus, the current work suggested the negative effects of HS on the ovarian tissue of female rabbits, and in conclusion these changes could be caused by decreased serum IL-2, IL-8, CAT and GSH-Px levels, increased ovarian apoptosis, and changed the expression of miRNAs.
Collapse
Affiliation(s)
- Lipeng Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xue Bai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiaohong Xie
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Guanhe Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ming Lei
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Congyan Li
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW This review aims to discuss the potential roles of omega-3 (ω-3) and omega-6 (ω-6) polyunsaturated fatty acids (PUFAs) in the prevention and treatment of metabolic diseases, to provide the latest evidence from epidemiological and clinical studies, and to highlight novel insights into this field. RECENT FINDINGS Higher dietary or circulating ω-3 PUFA levels are related to a lower risk of metabolic syndrome. Novel findings in obesity indicate higher proportions of ω-6 and ω-3 PUFAs, a modulated oxylipin profile and an altered transcriptome in subcutaneous white adipose tissue, that seem resistant to the effects of ω-3 PUFAs compared with what occurs in normal weight individuals. ω-3 PUFAs may improve the blood lipid profile and glycemic outcomes in patients with type 2 diabetes mellitus and reduce liver fat in nonalcoholic fatty liver disease (NAFLD); the findings of several recent meta-analyses support these effects. Genetic background affects inter-individual variability in the insulin sensitivity response to ω-3 PUFA supplementation. ω-3 PUFAs have prebiotic effects, altering the gut microbiota. SUMMARY Although evidence for health benefits of ω-3 PUFAs is strong, recent findings suggest a more personalized approach to ω-3 PUFA intake for individuals at high risk for metabolic diseases.
Collapse
Affiliation(s)
- Ivana Djuricic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
38
|
Brown JL, Peelor FF, Georgescu C, Wren JD, Kinter M, Tyrrell VJ, O'Donnell VB, Miller BF, Van Remmen H. Lipid hydroperoxides and oxylipins are mediators of denervation induced muscle atrophy. Redox Biol 2022; 57:102518. [PMID: 36283174 PMCID: PMC9593840 DOI: 10.1016/j.redox.2022.102518] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 01/14/2023] Open
Abstract
Loss of innervation is a key driver of age associated muscle atrophy and weakness (sarcopenia). Our laboratory has previously shown that denervation induced atrophy is associated with the generation of mitochondrial hydroperoxides and lipid mediators produced downstream of cPLA2 and 12/15 lipoxygenase (12/15-LOX). To define the pathological impact of lipid hydroperoxides generated in denervation-induced atrophy in vivo, we treated mice with liproxstatin-1, a lipid hydroperoxide scavenger. We treated adult male mice with 5 mg/kg liproxstain-1 or vehicle one day prior to sciatic nerve transection and daily for 7 days post-denervation before tissue analysis. Liproxstatin-1 treatment protected gastrocnemius mass and fiber cross sectional area (∼40% less atrophy post-denervation in treated versus untreated mice). Mitochondrial hydroperoxide generation was reduced 80% in vitro and by over 65% in vivo by liproxstatin-1 treatment in denervated permeabilized muscle fibers and decreased the content of 4-HNE by ∼25% post-denervation. Lipidomic analysis revealed detectable levels of 25 oxylipins in denervated gastrocnemius muscle and significantly increased levels for eight oxylipins that are generated by metabolism of fatty acids through 12/15-LOX. Liproxstatin-1 treatment reduced the level of three of the eight denervation-induced oxylipins, specifically 15-HEPE, 13-HOTrE and 17-HDOHE. Denervation elevated protein degradation rates in muscle and treatment with liproxstatin-1 reduced rates of protein breakdown in denervated muscle. In contrast, protein synthesis rates were unchanged by denervation. Targeted proteomics revealed a number of proteins with altered expression after denervation but no effect of liproxstain-1. Transcriptomic analysis revealed 203 differentially expressed genes in denervated muscle from vehicle or liproxstatin-1 treated mice, including ER stress, nitric oxide signaling, Gαi signaling, glucocorticoid receptor signaling, and other pathways. Overall, these data suggest lipid hydroperoxides and oxylipins are key drivers of increased protein breakdown and muscle loss associated with denervation induced atrophy and a potential target for sarcopenia intervention.
Collapse
Affiliation(s)
- Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Fredrick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Constantin Georgescu
- Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jonathan D Wren
- Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Victoria J Tyrrell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, CF14 4XN, United Kingdom
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, CF14 4XN, United Kingdom
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
39
|
Zachut M, Tam J, Contreras GA. Modulating immunometabolism in transition dairy cows: the role of inflammatory lipid mediators. Anim Front 2022; 12:37-45. [PMID: 36268169 PMCID: PMC9564993 DOI: 10.1093/af/vfac062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
| | - Joseph Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Genaro Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
40
|
Brace N, Megson IL, Rossi AG, Doherty MK, Whitfield PD. SILAC-based quantitative proteomics to investigate the eicosanoid associated inflammatory response in activated macrophages. J Inflamm (Lond) 2022; 19:12. [PMID: 36050729 PMCID: PMC9438320 DOI: 10.1186/s12950-022-00309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Macrophages play a central role in inflammation by phagocytosing invading pathogens, apoptotic cells and debris, as well as mediating repair of tissues damaged by trauma. In order to do this, these dynamic cells generate a variety of inflammatory mediators including eicosanoids such as prostaglandins, leukotrienes and hydroxyeicosatraenoic acids (HETEs) that are formed through the cyclooxygenase, lipoxygenase and cytochrome P450 pathways. The ability to examine the effects of eicosanoid production at the protein level is therefore critical to understanding the mechanisms associated with macrophage activation. RESULTS This study presents a stable isotope labelling with amino acids in cell culture (SILAC) -based proteomics strategy to quantify the changes in macrophage protein abundance following inflammatory stimulation with Kdo2-lipid A and ATP, with a focus on eicosanoid metabolism and regulation. Detailed gene ontology analysis, at the protein level, revealed several key pathways with a decrease in expression in response to macrophage activation, which included a promotion of macrophage polarisation and dynamic changes to energy requirements, transcription and translation. These findings suggest that, whilst there is evidence for the induction of a pro-inflammatory response in the form of prostaglandin secretion, there is also metabolic reprogramming along with a change in cell polarisation towards a reduced pro-inflammatory phenotype. CONCLUSIONS Advanced quantitative proteomics in conjunction with functional pathway network analysis is a useful tool to investigate the molecular pathways involved in inflammation.
Collapse
Affiliation(s)
- Nicole Brace
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Ian L Megson
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mary K Doherty
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Phillip D Whitfield
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK.
- Present Address: Glasgow Polyomics, Garscube Campus, University of Glasgow, Glasgow, G61 1BD, UK.
| |
Collapse
|
41
|
Angelotti A, Snoke DB, Ormiston K, Cole RM, Borkowski K, Newman JW, Orchard TS, Belury MA. Potential Cardioprotective Effects and Lipid Mediator Differences in Long-Chain Omega-3 Polyunsaturated Fatty Acid Supplemented Mice Given Chemotherapy. Metabolites 2022; 12:metabo12090782. [PMID: 36144189 PMCID: PMC9505633 DOI: 10.3390/metabo12090782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Many commonly used chemotherapies induce mitochondrial dysfunction in cardiac muscle, which leads to cardiotoxicity and heart failure later in life. Dietary long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) have demonstrated cardioprotective function in non-chemotherapy models of heart failure, potentially through the formation of LC n-3 PUFA-derived bioactive lipid metabolites. However, it is unknown whether dietary supplementation with LC n-3 PUFA can protect against chemotherapy-induced cardiotoxicity. To test this, 36 female ovariectomized C57BL/6J mice were randomized in a two-by-two factorial design to either a low (0 g/kg EPA + DHA) or high (12.2 g/kg EPA + DHA) LC n-3 PUFA diet, and received either two vehicle or two chemotherapy (9 mg/kg anthracycline + 90 mg/kg cyclophosphamide) tail vein injections separated by two weeks. Body weight and food intake were measured as well as heart gene expression and fatty acid composition. Heart mitochondria were isolated using differential centrifugation. Mitochondrial isolate oxylipin and N-acylethanolamide levels were measured by mass spectrometry after alkaline hydrolysis. LC n-3 PUFA supplementation attenuated some chemotherapy-induced differences (Myh7, Col3a1) in heart gene expression, and significantly altered various lipid species in cardiac mitochondrial preparations including several epoxy fatty acids [17(18)-EpETE] and N-acylethanolamines (arachidonoylethanolamine, AEA), suggesting a possible functional link between heart lipids and cardiotoxicity.
Collapse
Affiliation(s)
- Austin Angelotti
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Deena B. Snoke
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05405, USA
| | - Kate Ormiston
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rachel M. Cole
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - John W. Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
- Western Human Nutrition Research Center, United States Department of Agriculture-Agriculture Research Service, Davis, CA 95616, USA
- Department of Nutrition, University of California-Davis, Davis, CA 95616, USA
| | - Tonya S. Orchard
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Martha A. Belury
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Correspondence:
| |
Collapse
|
42
|
Hussain H, Vutipongsatorn K, Jiménez B, Antcliffe DB. Patient Stratification in Sepsis: Using Metabolomics to Detect Clinical Phenotypes, Sub-Phenotypes and Therapeutic Response. Metabolites 2022; 12:metabo12050376. [PMID: 35629881 PMCID: PMC9145582 DOI: 10.3390/metabo12050376] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Infections are common and need minimal treatment; however, occasionally, due to inappropriate immune response, they can develop into a life-threatening condition known as sepsis. Sepsis is a global concern with high morbidity and mortality. There has been little advancement in the treatment of sepsis, outside of antibiotics and supportive measures. Some of the difficulty in identifying novel therapies is the heterogeneity of the condition. Metabolic phenotyping has great potential for gaining understanding of this heterogeneity and how the metabolic fingerprints of patients with sepsis differ based on survival, organ dysfunction, disease severity, type of infection, treatment or causative organism. Moreover, metabolomics offers potential for patient stratification as metabolic profiles obtained from analytical platforms can reflect human individuality and phenotypic variation. This article reviews the most relevant metabolomic studies in sepsis and aims to provide an overview of the metabolic derangements in sepsis and how metabolic phenotyping has been used to identify sub-groups of patients with this condition. Finally, we consider the new avenues that metabolomics could open, exploring novel phenotypes and untangling the heterogeneity of sepsis, by looking at advances made in the field with other -omics technologies.
Collapse
Affiliation(s)
- Humma Hussain
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
| | - Kritchai Vutipongsatorn
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
| | - Beatriz Jiménez
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - David B. Antcliffe
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK; (H.H.); (K.V.)
- Correspondence:
| |
Collapse
|
43
|
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
|