1
|
Malhotra P, Fyfe J, Emmanouilidi A, Casari I, Mellett NA, Huynh K, Pajic M, Greening DW, Meikle PJ, Falasca M. Oncogenic small extracellular vesicles enriched in sphingosine-1-phosphate play a crucial role in pancreatic cancer progression. Cell Signal 2025; 132:111775. [PMID: 40158707 DOI: 10.1016/j.cellsig.2025.111775] [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: 01/13/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Small extracellular vesicles (sEVs) from tumour cells mediate intercellular communication and signalling to regulate the progression of pancreatic ductal adenocarcinoma (PDAC). While we and others have shown that PDAC-derived sEVs comprise oncogenic protein and nucleic acid cargo, understanding the lipid landscape of these sEVs remains unknown. Lipids influence both the composition of sEVs and their roles in lipid metabolism and signalling pathways within the tumour microenvironment and tumorigenesis. We hypothesised that specific lipids in oncogenic sEVs might provide insights into PDAC. Comprehensive mass spectrometry-based lipidomic analysis was performed using liquid chromatography-electrospray ionisation-tandem mass spectrometry on sEVs isolated from PDAC and non-malignant pancreatic cell lines, patient-derived xenograft cell lines and plasma from the PDAC transgenic mouse model KPC (KRASWT/G12D/ TP53WT/R172H/Pdx1-Cre+/+). The sEV lipidomic analyses identified over 700 lipid species from 25 lipid classes and subclasses. Our results showed that, compared to non-malignant cells, PDAC-derived sEVs were enriched in specific lysophospholipids, particularly sphingosine-1-phosphate (S1P), a lipid known for its pivotal role in cancer pathogenesis. S1P enrichment was validated in plasma-derived sEVs from KPC mice compared to WT. To explore the functional implications of S1P enrichment, we conducted assays demonstrating that S1P in sEVs facilitated tubule formation in human microvascular endothelial cells and promoted cancer-associated fibroblast cell migration. We show that PDAC-derived sEVs are differentially enriched in specific lipids associated with cancer phenotype. Our findings highlight that PDAC-derived sEVs are enriched in specific lipids, particularly S1P, which plays a crucial role in promoting cancer progression.
Collapse
Affiliation(s)
- Pratibha Malhotra
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Jordan Fyfe
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Aikaterini Emmanouilidi
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Ilaria Casari
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Marina Pajic
- Translational Oncology Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David W Greening
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
2
|
Petitfils C, Depommier C, Delzenne NM, Everard A, Van Hul M, Cani PD. Fecal Dysosmobacter spp. concentration is linked to plasma lipidome in insulin-resistant individuals with overweight, obesity and metabolic syndrome. Lipids Health Dis 2025; 24:203. [PMID: 40481506 PMCID: PMC12142885 DOI: 10.1186/s12944-025-02629-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/30/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Obesity is reaching epidemic proportions worldwide. This excessive increase of adipose tissue is a risk factor for the development of multiple diseases and premature death. Amongst associated diseases, metabolic syndrome is one of the main comorbidities of obesity. In this context, the gut microbiota has been recognized as both shaping and responding to host energy metabolism. Recently metabolomics has emerged as a powerful tool to capture a snapshot of the metabolites present in a specific tissue, providing new insights into host-microbiota interactions. Integrating metabolomics with gut microbiota studies could help us better understand how specific species impact on host metabolomic profile. Dysosmobacter welbionis has been identified as a promising next generation beneficial bacteria with potential effects on fat mass and glucose metabolism in mice, and fecal Dysosmobacter spp. concentration was inversely correlated to body mass index fasting glucose and plasmatic HbA1c in humans. METHODS Concentration of Dysosmobacter spp. was quantified by qPCR in the stools of insulin resistant overweight/obese participants with a metabolic syndrome and plasma metabolites were analyzed using untargeted metabolomics. Correlations between Dysosmobacter spp. fecal abundance and the 1169 identified plasma metabolites were uncovered using Spearman correlations followed by a false discovery rate correction. RESULTS Interestingly, among the detected metabolites, Dysosmobacter spp. was exclusively associated with lipid molecules. Fecal concentration of Dysosmobacter spp. was positively associated with plasmatic levels of five phosphatidylcholines, arachidonate, two monoacylglycerols, twelve diacylglycerols, three lysophosphatidylethanolamines, one phosphatidylinositol and three lysophosphatidylinositols, as well as glycerophosphoethanolamines, glycerophosphatidylcholine and PC(P-16:0). The correlation was particularly interesting with acylcholine and lysophosphatidylcholine metabolites as, respectively, 6/8 and 8/10 detected molecules were positively associated with Dysosmobacter spp. CONCLUSION These results suggest that Dysosmobacter spp. plays a specific role in host lipid metabolism. This finding aligns with previous in vivo studies highlighting lipid profile alterations in multiple tissues of mice treated with this bacterium. Further studies are needed to elucidate the underlying mechanisms and assess its potential therapeutic applications.
Collapse
Affiliation(s)
- Camille Petitfils
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Clara Depommier
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium.
| | - Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
3
|
Surendran A, Zhang H, Stamenkovic A, Ravandi A. Lipidomics and cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167806. [PMID: 40122185 DOI: 10.1016/j.bbadis.2025.167806] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide, necessitating innovative approaches for early detection and personalized interventions. Lipidomics, leveraging advanced mass spectrometry techniques, has become instrumental in deciphering lipid-mediated mechanisms in CVDs. This review explores the application of lipidomics in identifying biomarkers for myocardial infarction, heart failure, stroke, and calcific aortic valve stenosis (CAVS). This review examines the technological advancements in shotgun lipidomics and LC/MS, which provide unparalleled insights into lipid composition and function. Key lipid biomarkers, including ceramides and lysophospholipids, have been linked to disease progression and therapeutic outcomes. Integrating lipidomics with genomic and proteomic data reveals the molecular underpinnings of CVDs, enhancing risk prediction and intervention strategies. This review positions lipidomics as a transformative tool in reshaping cardiovascular research and clinical practice.
Collapse
Affiliation(s)
- Arun Surendran
- Mass Spectrometry Core Facility, BRIC-Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Hannah Zhang
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada
| | - Aleksandra Stamenkovic
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada
| | - Amir Ravandi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada.
| |
Collapse
|
4
|
Mondal S, Pandey U, Chakrabarti S, Pahchan P, Koner D, Banerjee S. Rapid and Reagent-Free Analysis of Dried Blood Spot by Paper Spray Mass Spectrometry Reveals Sex: Implications in Forensic Investigations. J Proteome Res 2025; 24:2314-2323. [PMID: 39842085 DOI: 10.1021/acs.jproteome.4c00798] [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: 01/24/2025]
Abstract
Identifying sex from an unknown dried blood spot (DBS), especially when the corpse remains undiscovered, often provides valuable evidence in forensic casework. While DNA-based sex determination is a reliable method in forensic settings, it requires expensive reagents and is time-consuming. To develop a rapid reagent-free blood test for sex, we employed paper spray ionization mass spectrometry (PSI-MS) to capture sex-discriminatory lipid profiles from 200 DBS samples comprising 100 males and 100 females. We conducted a supervised machine learning (ML) analysis on all detected lipid signals to hunt biomarkers of sex within the data set. This analysis unveiled significant differences in specific sphingomyelin and phospholipid species levels between male and female DBS samples. Using the parsimonious set of 60 lipid signals, we constructed a classifier that achieved 100% overall accuracy in predicting sex from DBS on paper. Additionally, we assessed three-day-old air-exposed DBS on glass and granite surfaces, simulating the typical blood samples available for forensic investigations. Consequently, we achieved ∼92% overall sex prediction accuracy from the holdout test data set of DBS on glass and granite surfaces. As a highly sensitive detection tool, PSI-MS combined with ML has the potential to revolutionize forensic methods by rapidly analyzing blood molecules encoding personal information.
Collapse
Affiliation(s)
- Supratim Mondal
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| | - Uddeshya Pandey
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| | - Sourik Chakrabarti
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| | - Pragya Pahchan
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| | - Debasish Koner
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Shibdas Banerjee
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati 517507, India
| |
Collapse
|
5
|
Paul S, Morgan P, Pernes G, Schooneveldt Y, Duong T, Mellett NA, Huynh K, Murphy AJ, Lancaster GI, Meikle PJ. Modulation of endogenous plasmalogens by genetic ablation of lysoplasmalogenase (Tmem86b) in mice. J Lipid Res 2025; 66:100808. [PMID: 40245986 PMCID: PMC12144449 DOI: 10.1016/j.jlr.2025.100808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
Plasmalogens are a distinct subclass of glycerophospholipids that exhibit unique structural features, notably possessing a vinyl ether linkage at the sn1 position of the glycerol backbone. These specialized lipids play crucial roles in various biological functions. Although the biosynthetic pathway of plasmalogens has been well-characterized, their catabolism remains less studied. In this study, we investigated the impact of global and tissue-specific loss-of-function of a plasmalogen catabolizing enzyme, lysoplasmalogenase (TMEM86B), on circulatory and tissue lipidomes. We generated both global and hepatocyte-specific Tmem86b knockout mice using cre-loxP technology. Mice with homozygous global inactivation of Tmem86b (Tmem86b KO mice) were viable and did not display any marked phenotypic abnormalities. Tmem86b KO mice demonstrated significantly elevated levels of the plasmalogens, alkenylphosphatidylethanolamine (PE(P)), and alkenylphosphatidylcholine (PC(P)), as well as lysoplasmalogens, in the plasma, liver, and natural killer cells compared to their wild-type counterparts. The endogenous alkenyl chain composition of plasmalogens remained unaltered in Tmem86b KO mice. Consistent with the global knockout findings, hepatocyte-specific Tmem86b knockout mice also exhibited increased plasmalogen levels in the plasma and liver compared to their floxed control counterparts. Overall, our findings shed light on the role of Tmem86b in plasmalogen catabolism, demonstrating how its ablation leads to elevated plasmalogen levels in select tissues and cells. This study enhances our understanding of the regulatory mechanisms governing plasmalogen metabolism and highlights the potential of targeting Tmem86b to therapeutically raise plasmalogen levels.
Collapse
Affiliation(s)
- Sudip Paul
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia.
| | - Pooranee Morgan
- Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gerard Pernes
- Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yvette Schooneveldt
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Thy Duong
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew J Murphy
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Graeme I Lancaster
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Varghese M, Thekkelnaycke R, Soni T, Zhang J, Maddipati K, Singer K. Sex differences in the lipid profiles of visceral adipose tissue with obesity and gonadectomy. J Lipid Res 2025; 66:100803. [PMID: 40245983 PMCID: PMC12144442 DOI: 10.1016/j.jlr.2025.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/05/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
In obesity, adipose tissue (AT) expansion is accompanied by chronic inflammation. Altered lipid composition in the visceral or gonadal white AT (GWAT) directly drive AT macrophage accumulation and activation to a proinflammatory phenotype. Sex steroid hormones modulate visceral versus subcutaneous lipid accumulation that correlates with metabolic syndrome, especially in men and postmenopausal women who are more prone to abdominal obesity. Prior studies demonstrated sex differences in GWAT lipid species in HFD-fed mice, but the role of sex hormones is still unclear. We hypothesized that sex hormone alterations with gonadectomy (GX) would further impact lipid composition in the obese GWAT. Untargeted lipidomics of obese GWAT identified sex differences in phospholipids, sphingolipids, sterols, fatty acyls, saccharolipids and prenol lipids. Males had significantly more precursor fatty acids (palmitic, oleic, linoleic, and arachidonic acid) than females and GX mice. Targeted lipidomics for fatty acids and oxylipins in the HFD-fed male and female GWAT stromal vascular fraction identified higher omega-6 to omega-3 free fatty acid profile in males and differences in PUFAs-derived prostaglandins, thromboxanes, and leukotrienes. Both obese male and female GWAT stromal vascular fraction showed increased levels of arachidonic acid-derived oxylipins compared to their lean counterparts. Bulk RNA-seq of sorted GWAT AT macrophages highlighted sex and diet differences in PUFA and oxylipin metabolism genes. These findings of sexual dimorphism in both stored lipid species and PUFA-derived mediators with diet and GX emphasize sex differences in lipid metabolism pathways that drive inflammation responses and metabolic disease risk in obesity.
Collapse
Affiliation(s)
- Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rajendiran Thekkelnaycke
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Jiayu Zhang
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | | | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Shi M, Li W, Yang S, Lv Q, Yang J, Sun D, Yang G, Zhao Y, Zhang W, Li M, Yang Y, Cai C, Gao P, Guo X, Li B, Cao G. Integrated Multi-Tissue Lipidomics and Transcriptomics Reveal Differences in Lipid Composition Between Mashen and Duroc × (Landrace × Yorkshire) Pigs. Animals (Basel) 2025; 15:1280. [PMID: 40362094 PMCID: PMC12071155 DOI: 10.3390/ani15091280] [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: 03/03/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Chinese native pig breeds exhibit unique advantages over Western pig breeds, but the specific lipid metabolism mechanisms remain unclear. The phenotypic characteristics of Mashen (MS) pigs and Duroc × (Landrace × Yorkshire) (DLY) pigs are studied. The results show that MS pigs exhibit higher intramuscular fat (IMF) content. The area of adipocytes of MS pigs is significantly greater than that in DLY pigs (p < 0.01). Lipidomics analysis reveals distinct profiles in the upper layer of backfat (ULB), leaf lard (LL), greater omentum (GOM), and IMF, with MS pigs showing higher polyunsaturated fatty acids (PUFAs) in ULB, LL, and GOM. Key differential lipids identified in the two pig breeds include the following triglycerides (TGs) and phosphatidylcholines (PC): TG(16:1_18:1_18:3), TG(18:1_18:2_18:3), TG(18:3_18:2_18:2), PC(18:0_18:1), and PC(18:0_18:2). Weighted gene co-expression network analysis (WGCNA) reveals lipid molecules associated with serum biochemical indices. Transcriptomics analysis highlights 1944 differentially expressed genes between the MS-ULB and DLY-ULB. Notably, multiple genes from the cytochrome P450 family (CYP2E1, CYP4A24, CYP2J2), along with PLA2G2D, PLA2G4A, and multiple PCs, are associated with the metabolism of arachidonic acids and linoleic acids. PLA2G2D and PLA2G4A are also involved in the metabolism of α-linolenic acids. This comprehensive analysis provides essential information for breeding strategies and meat quality improvement.
Collapse
Affiliation(s)
- Mingyue Shi
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Wenxia Li
- Institute of Ecological Agriculture and Animal Husbandry, Shanxi Agricultural University, Shuozhou 036002, China;
| | - Shuai Yang
- Shanxi Animal Husbandry Technology Extension Service Center, Taiyuan 030001, China;
| | - Qipin Lv
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Jingxian Yang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Di Sun
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Guanqing Yang
- Taigu Modern Agricultural Industry Development Center of Jinzhong City, Jinzhong 030801, China;
| | - Yan Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Wanfeng Zhang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Meng Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Yang Yang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Pengfei Gao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (M.S.); (Q.L.); (J.Y.); (D.S.); (Y.Z.); (W.Z.); (M.L.); (Y.Y.); (C.C.); (P.G.); (X.G.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| |
Collapse
|
8
|
Kalkman HO, Smigielski L. Ceramides may Play a Central Role in the Pathogenesis of Alzheimer's Disease: a Review of Evidence and Horizons for Discovery. Mol Neurobiol 2025:10.1007/s12035-025-04989-0. [PMID: 40295359 DOI: 10.1007/s12035-025-04989-0] [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: 02/06/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
While several hypotheses have been proposed to explain the underlying mechanisms of Alzheimer's disease, none have been entirely satisfactory. Both genetic and non-genetic risk factors, such as infections, metabolic disorders and psychological stress, contribute to this debilitating disease. Multiple lines of evidence indicate that ceramides may be central to the pathogenesis of Alzheimer's disease. Tumor necrosis factor-α, saturated fatty acids and cortisol elevate the brain levels of ceramides, while genetic risk factors, such as mutations in APP, presenilin, TREM2 and APOE ε4, also elevate ceramide synthesis. Importantly, ceramides displace sphingomyelin and cholesterol from lipid raft-like membrane patches that connect the endoplasmic reticulum and mitochondria, disturbing mitochondrial oxidative phosphorylation and energy production. As a consequence, the flattening of lipid rafts alters the function of γ-secretase, leading to increased production of Aβ42. Moreover, ceramides inhibit the insulin-signaling cascade via at least three mechanisms, resulting in the activation of glycogen synthase kinase-3 β. Activation of this kinase has multiple consequences, as it further deteriorates insulin resistance, promotes the transcription of BACE1, causes hyperphosphorylation of tau and inhibits the transcription factor Nrf2. Functional Nrf2 prevents apoptosis, mediates anti-inflammatory activity and improves blood-brain barrier function. Thus, various seemingly unrelated Alzheimer's disease risk factors converge on ceramide production, whereas the elevated levels of ceramides give rise to the well-known pathological features of Alzheimer's disease. Understanding and targeting these mechanisms may provide a promising foundation for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Hans O Kalkman
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Lukasz Smigielski
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Wu W, Huynh K, Du JC, She G, Duong T, Ziemann M, Zhao WB, Deng XL, Meikle PJ, Du XJ. Hippo pathway activation causes multiple lipid derangements in a murine model of cardiomyopathy. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159590. [PMID: 39709046 DOI: 10.1016/j.bbalip.2024.159590] [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: 07/19/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Metabolic reprogramming occurs in cardiomyopathy and heart failure contributing to progression of the disease. Activation of cardiac Hippo pathway signaling has been implicated in mediating mitochondrial dysfunction and metabolic reprogramming in cardiomyopathy, albeit influence of Hippo pathway on lipid profile is unclear. Using a dual-omics approach, we determined alterations of cardiac lipids in a mouse model of cardiomyopathy due to enhanced Hippo signaling and explored molecular mechanisms. Lipidomic profiling discovered multiple alterations in lipid classes, notably reduction of triacylglycerol, diacylglycerol, phospholipids and ether lipids, and elevation of sphingolipids and lysophosphatidylcholine. Mechanistically, we found downregulated expression of PPARα and PGC-1α at mRNA and protein levels, and downregulated expression of PPARα-target genes, indicating attenuated transcriptional activity of PPARα/PGC-1α. Lipidomics-guided transcriptomic analysis revealed dysregulated expression of gene sets that were responsible for enhanced biosynthesis of ceramides, suppression of TG biosynthesis, storage, hydrolysis and mitochondrial fatty acid oxidation, and reduction of peroxisome-localized biosynthesis of ether lipids. Collectively, Hippo pathway activation with attenuated PPARα/PGC-1α signaling is the underlying mechanism for alterations in cardiac lipids in cardiomyopathy and failing heart.
Collapse
Affiliation(s)
- Wei Wu
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jin-Chan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia; Bioinformatics Working Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Endocrinology, The Ninth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Xiao-Jun Du
- Department of Cardiology, Shaanxi Provincial Hospital, Xi'an, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
da Silva Rosa SC, Alizadeh J, Vitorino R, Surendran A, Ravandi A, Kidane B, Ghavami S. A Lipidomics Approach to Determine the Role of Lipids and Its Crosstalk with Autophagy in Lung Cancer Metastasis. Methods Mol Biol 2025; 2879:239-260. [PMID: 38441721 DOI: 10.1007/7651_2024_524] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
Non-small cell lung cancer (NSCLC) is among the most malignant tumors with high propensity for metastasis and is the leading cause of cancer-related death globally. Most patients present with regional and distant metastasis, associated with poor prognosis. Lipids may play an essential role in either activating or inhibiting detachment-induced apoptosis (anoikis), where the latter is a crucial mechanism to prevent metastasis, and it may have a cross-talk with autophagy. Autophagy has been shown to be induced in various human cancer metastasis, modulating tumor cell motility and invasion, cancer cell differentiation, resistance to anoikis, and epithelial to mesenchymal transition. Hence, it may play a crucial role in the transition of benign to malignant phenotypes, the core of metastasis initiation. Here, we provide a method we have established in our laboratory for detecting lipids in attached and detached non-small lung cancer cells and show how to analyze lipidomics data to find its correlation with autophagy-related pathways.
Collapse
Affiliation(s)
- Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rui Vitorino
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, Aveiro, Portugal
| | - Arun Surendran
- Mass Spectrometry Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Biniam Kidane
- Department of Surgery, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
11
|
Zhao S, Giles C, Huynh K, Kettunen J, Järvelin MR, Kähönen M, Viikari J, Lehtimäki T, Raitakari OT, Meikle PJ, Mäkinen VP, Ala-Korpela M. Personalized Profiling of Lipoprotein and Lipid Metabolism Based on 1018 Measures from Combined Quantitative NMR and LC-MS/MS Platforms. Anal Chem 2024; 96:20362-20370. [PMID: 39680883 PMCID: PMC11696825 DOI: 10.1021/acs.analchem.4c03229] [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: 06/24/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Applications of advanced omics methodologies are increasingly popular in biomedicine. However, large-scale studies aiming at clinical translation are typically siloed to single technologies. Here, we present the first comprehensive large-scale population data combining 209 lipoprotein measures from a quantitative NMR spectroscopy platform and 809 lipid classes and species from a quantitative LC-MS/MS platform. These data with 1018 molecular measures were analyzed in two population cohorts totaling 7830 participants. The association and cluster analyses revealed excellent coherence between the methodologically independent data domains and confirmed their quantitative compatibility and suitability for large-scale studies. The analyses elucidated the detailed molecular characteristics of the heterogeneous circulatory macromolecular lipid transport system and the underlying structural and compositional relationships. Unsupervised neural network analysis─the so-called self-organizing maps (SOMs)─revealed that these deep molecular and metabolic data are inherently related to key physiological and clinical population characteristics. The data-driven population subgroups uncovered marked differences in the population distribution of multiple cardiometabolic risk factors. These include, e.g., multiple lipoprotein lipids, apolipoprotein B, ceramides, and oxidized lipids. All 79 structurally unique triglyceride species showed similar associations over the entire lipoprotein cascade and indicated systematically increased risk for carotid intima media thickening and other atherosclerosis risk factors, including obesity and inflammation. The metabolic attributes for 27 individual cholesteryl ester species, which formed six distinct clusters, were more intricate with associations both with higher─e.g., CE(16:1)─and lower─e.g., CE(20:4)─cardiometabolic risk. The molecular details provided by these combined data are unprecedented for molecular epidemiology and demonstrate a new potential avenue for population studies.
Collapse
Affiliation(s)
- Siyu Zhao
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
| | - Corey Giles
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
| | - Kevin Huynh
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
| | - Johannes Kettunen
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
- Department
of Public Health and Welfare, Finnish Institute
for Health and Welfare, 00271 Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Department
of Epidemiology and Biostatistics, MRC Centre for Environment and
Health, School of Public Health, Imperial
College London, London W12 0BZ, U.K.
- Department
of Life Sciences, College of Health and Life Sciences, Brunel University London, London UB8 3PH, U.K.
| | - Mika Kähönen
- Department
of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular
Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, 33270 Tampere, Finland
| | - Jorma Viikari
- Department
of Medicine, University of Turku, 20014 Turku, Finland
- Division
of Medicine, Turku University Hospital, 20014 Turku, Finland
| | - Terho Lehtimäki
- Department
of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular
Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, 33270 Tampere, Finland
| | - Olli T. Raitakari
- Research
Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20014 Turku, Finland
- Centre
for Population Health Research, University
of Turku and Turku University Hospital, 20014 Turku, Finland
- Department
of Clinical Physiology and Nuclear Medicine, Turku University Hospital, 20014 Turku, Finland
- InFLAMES
Research Flagship, University of Turku, 20014 Turku, Finland
| | - Peter J. Meikle
- Baker
Heart and Diabetes Institute, Melbourne 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Melbourne 3004, Australia
- Monash
University, Melbourne 3004, Australia
| | - Ville-Petteri Mäkinen
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
| | - Mika Ala-Korpela
- Systems
Epidemiology, Faculty of Medicine, University
of Oulu, 90014 Oulu, Finland
- Research
Unit of Population Health, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter
Oulu, 90014 Oulu, Finland
- Monash
University, Melbourne 3004, Australia
- NMR Metabolomics
Laboratory, School of Pharmacy, University
of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
12
|
Zhang S, Zhai Z, Gao T, Kuai X, Li X, Dong Y, Lu C, Zhuo K, Xiang Q, Liu D. Identification of serum metabolic traits of AIWG in first-episode schizophrenia patients. BMC Psychiatry 2024; 24:946. [PMID: 39716136 PMCID: PMC11667919 DOI: 10.1186/s12888-024-06413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Antipsychotic-induced weight gain (AIWG) is a common side effect of antipsychotic drugs and may lead to cardiometabolic comorbidities. There is an urgent public health need to identify patients at high risk of AIWG and determine potential biomarkers for AIWG. METHODS In the Sequential Multiple-Assignment Randomized Trials to Compare Antipsychotic Treatments (SMART-CAT) trail, first-episode schizophrenia patients were randomly assigned to olanzapine, risperidone, perphenazine, amisulpride or aripiprazole for 8 weeks. We applied absolute quantitative lipidomics at baseline and after 8 weeks of antipsychotic treatment in 80 patients. To evaluate the effects of AIWG on lipid profile, 25 patients with ≥ 7% weight changes (weight gain, WG) and 28 patients with <|3|% weight changes (weight stable, WS) were investigated, separately. RESULTS We found that baseline CerP(d40:3) and PC(20:1_22:6) were positively associated with weight changes at follow-up (r > 0.4, pFDR < 0.05). Additionally, baseline CerP(d40:3) and PC(20:1_22:6) independently predicted rapid weight gain, with receiver operating curve (ROC) of 0.76 (95% CI: 0.63-0.90), and 0.75 (95% CI: 0.62-0.88), respectively. Compared with baseline, levels of 45 differential lipid metabolites (fold change > 1.2, VIP > 1 and pFDR < 0.05) were significantly higher in the WG group. Interestingly, no differential lipid metabolites were identified in the WS group. The LASSO regression model identified 18 AIWG lipid signatures, including 2 cholesterol esters (ChEs), 1 diglyceride (DG), 12 phosphatidylcholines (PCs), 1 phosphatidylglycerol (PG), 1 phosphatidylinositol (PI), and 1 sphingomyelin (SM), with the ChE(16:1) contributing the most. Furthermore, the level changes of ChE(16:1) were positively associated with weight gain(r = 0.67, pFDR < 0.05). CONCLUSION Our findings indicate that lipid profile may serve as predictors of rapid weight gain in schizophrenia and provide useful markers for AIWG intervention.
Collapse
Affiliation(s)
- Suzhen Zhang
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
| | - Zhaolin Zhai
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianhao Gao
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinping Kuai
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
| | - Xuan Li
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
| | - Yuke Dong
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Chang Lu
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Kaiming Zhuo
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China
| | - Qiong Xiang
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China.
| | - Dengtang Liu
- Division of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Clinical Center for Psychotic Disorders, National Center for Mental Disorders, Shanghai, China.
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China.
- Institute of Mental Health, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Hahnefeld L, Hackel J, Trautmann S, Angioni C, Schreiber Y, Gurke R, Thomas D, Wicker S, Geisslinger G, Tegeder I. Healthy plasma lipidomic signatures depend on sex, age, body mass index, and contraceptives but not perceived stress. Am J Physiol Cell Physiol 2024; 327:C1462-C1480. [PMID: 39437447 DOI: 10.1152/ajpcell.00630.2024] [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: 08/30/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Perceived stress is thought to contribute to the pathogenesis of metabolic, vascular, mental, and immune diseases, with different susceptibilities in women and men. The present study investigated if and how perceived stress and/or demographic variables, including sex, age, body mass index, regular prescription drugs, occasional analgesics, or dietary supplements, manifested in plasma lipidomic profiles obtained by targeted and untargeted mass spectrometry analyses. The study included 217 healthy women and 108 healthy men, aged 18-68 yr, who were recruited in a 2:1 female:male ratio to account for women with/without contraceptives. As expected, dehydroepiandrosterone sulfate (DHEAS) and ceramides were higher in men than women, and DHEAS decreased with age, whereas ceramides increased. Contrary to expectations, neither DHEAS nor ceramides were associated with perceived stress [Perceived Stress Questionnaire with 30 questions (PSQ30 questionnaire)], which was, however, associated with BMI in men but not in women. None of the lipid species or classes showed a similar "age × sex × BMI" interaction, but the endocannabinoid palmitoylethanolamide (PEA) correlated with body mass index (BMI) and hypertension. Independent of perceived stress, lysophosphatidylcholines (LPCs) were lower in women than men, whereas LPC metabolites, lysophosphatidic acids (LPAs), were higher in women. The LPA:LPC ratio was particularly high in women using oral contraceptives, suggesting a strong hormone-induced extracellular conversion of LPCs to LPAs, which is catalyzed by the phospholipase D, autotaxin. The results reveal complex sex differences in perceived stress and lipidomic profiles, the latter being exacerbated by contraceptive use, but perceived stress and lipids were not directly correlated.NEW & NOTEWORTHY Perceived stress (PSQ questionnaire) depends on the interaction of "sex × age × BMI." Plasma lipid profiles depend on sex and age. Natural sex differences are exacerbated by the use of contraceptives. Perceived stress is not correlated with specific plasma lipids or lipidomic profiles. Women have high LPA:LPC ratios in association with high levels of autotaxin.
Collapse
Affiliation(s)
- Lisa Hahnefeld
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Juliane Hackel
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sandra Trautmann
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Carlo Angioni
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Robert Gurke
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Dominique Thomas
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Sabine Wicker
- Goethe-University Frankfurt, University Hospital, Occupational Health Service, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Keating MF, Yang C, Liu Y, Gould EA, Hallam MT, Henstridge DC, Mellett NA, Meikle PJ, Watt KI, Gregorevic P, Calkin AC, Drew BG. Hepatic retinol dehydrogenase 11 dampens stress associated with the maintenance of cellular cholesterol levels. Mol Metab 2024; 90:102041. [PMID: 39362601 PMCID: PMC11752124 DOI: 10.1016/j.molmet.2024.102041] [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: 07/09/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVE Dysregulation of hepatic cholesterol metabolism can contribute to elevated circulating cholesterol levels, which is a significant risk factor for cardiovascular disease. Cholesterol homeostasis in mammalian cells is tightly regulated by an integrated network of transcriptional and post-transcriptional signalling pathways. Whilst prior studies have identified many of the central regulators of these pathways, the extended supporting networks remain to be fully elucidated. METHODS Here, we leveraged an integrated discovery platform, combining multi-omics data from 107 strains of mice to investigate these supporting networks. We identified retinol dehydrogenase 11 (RDH11; also known as SCALD) as a novel protein associated with cholesterol metabolism. Prior studies have suggested that RDH11 may be regulated by alterations in cellular cholesterol status, but its specific roles in this pathway are mostly unknown. RESULTS Here, we show that mice fed a Western diet (high fat, high cholesterol) exhibited a significant reduction in hepatic Rdh11 mRNA expression. Conversely, mice treated with a statin (3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) inhibitor) exhibited a 2-fold increase in hepatic Rdh11 mRNA expression. Studies in human and mouse hepatocytes demonstrated that RDH11 expression was regulated by altered cellular cholesterol conditions in a manner consistent with SREBP2 target genes HMGCR and LDLR. Modulation of RDH11 in vitro and in vivo demonstrated modulation of pathways associated with cholesterol metabolism, inflammation and cellular stress. Finally, RDH11 silencing in mouse liver was associated with a reduction in hepatic cardiolipin abundance and a concomitant reduction in the abundance of proteins of the mitochondrial electron transport chain. CONCLUSION Taken together, these findings suggest that RDH11 likely plays a role in protecting cells against the cellular toxicity that can arise as a by-product of endogenous cellular cholesterol synthesis.
Collapse
Affiliation(s)
- Michael F Keating
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Disease, University of Melbourne, Melbourne, Victoria, Australia
| | - Christine Yang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yingying Liu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Eleanor Am Gould
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mitchell T Hallam
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Darren C Henstridge
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Disease, University of Melbourne, Melbourne, Victoria, Australia; Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| | - Kevin I Watt
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia; Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia; Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Disease, University of Melbourne, Melbourne, Victoria, Australia; Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Disease, University of Melbourne, Melbourne, Victoria, Australia; Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
15
|
López-Yerena A, Muñoz-García N, de Santisteban Villaplana V, Padro T, Badimon L. Effect of Moderate Beer Intake on the Lipid Composition of Human Red Blood Cell Membranes. Nutrients 2024; 16:3541. [PMID: 39458535 PMCID: PMC11510343 DOI: 10.3390/nu16203541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Growing evidence suggests that erythrocyte membrane lipids are subject to changes during their lifespan. Factors such as the type of dietary intake and its composition contribute to the changes in red blood cell (RBC) membranes. Due to the high antioxidant content of beer, we aimed to investigate the effect of moderate beer consumption on the lipid composition of RBCs membranes from healthy overweight individuals. Methods: We conducted a four-weeks, prospective two-arm longitudinal crossed-over study, where participants (n = 36) were randomly assigned to alcohol-free beer group or traditional beer group. The lipids of RBCs membranes were assessed at the beginning and the end of the intervention by thin-layer chromatography. Results: Four-weeks of alcohol-free beer promoted changes in fatty acids (FA), free cholesterol (FC), phosphatidylethanolamine (PE) and phosphatidylcholine (PC) (p < 0.05). Meanwhile, traditional beer intake led to changes in FA, FC, phospholipids (PL), PE and PC (p < 0.05). The observed alterations in membrane lipids were found to be independent of sex and BMI as influencing factors. Conclusions: The lipid composition of erythrocyte membranes is distinctly but mildly influenced by the consumption of both non-alcoholic and conventional beer, with no effects on RBC membrane fluidity.
Collapse
Affiliation(s)
- Anallely López-Yerena
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
| | - Natalia Muñoz-García
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
| | - Victoria de Santisteban Villaplana
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- School of Pharmacy and Food Sciences, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Teresa Padro
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Lina Badimon
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| |
Collapse
|
16
|
Wilkin C, Piette J, Legrand-Poels S. Unravelling metabolic factors impacting iNKT cell biology in obesity. Biochem Pharmacol 2024; 228:116436. [PMID: 39029630 DOI: 10.1016/j.bcp.2024.116436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Obesity and related diseases have reached epidemic proportions and continue to rise. Beyond creating an economical burden, obesity and its co-morbidities are associated with shortened human life expectancy. Despite major advances, the underlying mechanisms of obesity remain not fully elucidated. Recently, several studies have highlighted that various immune cells are metabolically reprogrammed in obesity, thereby profoundly affecting the immune system. This sheds light on a new field of interest: the impact of obesity-related systemic metabolic changes affecting immune system that could lead to immunosurveillance loss. Among immune cells altered by obesity, invariant Natural Killer T (iNKT) cells have recently garnered intense focus due to their ability to recognize lipid antigen. While iNKT cells are well-described to be affected by obesity, how and to what extent immunometabolic factors (e.g., lipids, glucose, cytokines, adipokines, insulin and free fatty acids) can drive iNKT cells alterations remains unclear, but represent an emerging field of research. Here, we review the current knowledge on iNKT cells in obesity and discuss the immunometabolic factors that could modulate their phenotype and activity.
Collapse
Affiliation(s)
- Chloé Wilkin
- Laboratory of Immunometabolism and Nutrition, GIGA, ULiège, Liège, Belgium.
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA, ULiège, Liège, Belgium
| | | |
Collapse
|
17
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
18
|
Sourianarayanane A, Brydges CR, McCullough AJ. Liver tissue lipids in metabolic dysfunction-associated steatotic liver disease with diabetes and obesity. Clin Res Hepatol Gastroenterol 2024; 48:102402. [PMID: 38909684 DOI: 10.1016/j.clinre.2024.102402] [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: 04/18/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Diabetes and obesity are associated with altered lipid metabolism and hepatic steatosis. Studies suggest that increases in lipid accumulation in these patients with metabolic dysfunction-associated steatotic liver disease (MASLD) are not uniform for all lipid components. This study evaluates this variation. METHODS A comprehensive lipidomic analysis of different lipid groups, were performed on liver tissue and plasma samples obtained at the time of histology from a well-defined cohort of 72 MASLD participants. The lipid profiles of controls were compared to those of MASLD patients with obesity, diabetes, or a combination of both. RESULTS MASLD patients without obesity or diabetes exhibited distinct changes in the lipid profile of their liver tissue. The presence of diabetes or obesity further modified these lipid profiles (e.g., ceramide 47:7;4O), with positive or negative correlation (p < 0.05). A step-wise increase (long-chain fatty acids, triglycerides, and ceramides) or decrease (ultra-long fatty acids, diglycerides, and phospholipids) for lipid groups was observed compared to control among patients with MASLD without obesity or diabetes to MASLD patients with obesity as a single risk factor, and MASLD patients with obesity and diabetes. Changes in lipids observed in the plasma did not align with their corresponding liver tissue findings. CONCLUSION The changes observed in the composition of lipids are not similar in patients with obesity and diabetes among those with MASLD. This highlights the different metabolic processes at play. The presence of obesity or diabetes in patients with MASLD exacerbates these lipid derangements, underscoring the potential for targeted intervention in MASLD patients.
Collapse
Affiliation(s)
- Achuthan Sourianarayanane
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | |
Collapse
|
19
|
Brandao CFC, Krempf M, Giolo de Carvalho F, Aguesse A, Junqueira-Franco MVM, Batitucci G, de Freitas EC, Noronha NY, Rodrigues GDS, Junqueira GP, Borba DA, Billon-Crossouard S, Croyal M, Marchini JS. Sphingolipid and Trimethylamine-N-Oxide (TMAO) Levels in Women with Obesity after Combined Physical Training. Metabolites 2024; 14:398. [PMID: 39195494 DOI: 10.3390/metabo14080398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 08/29/2024] Open
Abstract
Obesity causes metabolic changes, such as the development of cardiovascular diseases. Moreover, physical exercise promotes protection against these diseases. Thus, the objective of the present study was to evaluate whether combined physical training can improve the metabolic system of women with obesity, reducing plasma concentrations of trimethylamine N-oxide (TMAO) and sphingolipids, regardless of weight loss. Fourteen obese women (BMI 30-40 kg/m2), aged 20-40 years, sedentary, were submitted to 8 weeks of combined physical training (strength and aerobic exercises). The training was performed three times/week, 55 min/session, at 75-90% maximum heart rate. All participants were evaluated pre- and post-exercise intervention, and their body composition, plasma TMAO, creatinine, lipid profile, and sphingolipid concentrations were recorded. Maximum oxygen consumption (VO2max), Speed lactate threshold 1 (SpeedLT1), and Speed lactate threshold 2 (SpeedLT2) evaluated physical performance. Results: After combined exercise, it did not change body composition, but TMAO, total cholesterol, and sphingolipid concentrations significantly decreased (p < 0.05). There was an increase in physical performance by improving VO2max, SpeedLT1, and SpeedLT2 (p < 0.05). The combined physical exercise could induce cardiovascular risk protection by decreasing TMAO in obese women, parallel to physical performance improvement, independent of weight loss.
Collapse
Affiliation(s)
- Camila Fernanda Cunha Brandao
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
- Department of Physical Education, State University of Minas Gerais, Divinópolis 35500-000, Minas Gerais, Brazil
| | - Michel Krempf
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Flávia Giolo de Carvalho
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Audrey Aguesse
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | | | - Gabriela Batitucci
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of São Paulo, Rod. Araraquara-Jau Km 1, Araraquara 14800-000, São Paulo, Brazil
| | - Ellen Cristini de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of São Paulo, Rod. Araraquara-Jau Km 1, Araraquara 14800-000, São Paulo, Brazil
| | - Natalia Yumi Noronha
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Guilherme da Silva Rodrigues
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Gizela Pedroso Junqueira
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| | - Diego Alcantara Borba
- Department of Physical Education, State University of Minas Gerais, Divinópolis 35500-000, Minas Gerais, Brazil
| | - Stéphanie Billon-Crossouard
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Mikael Croyal
- NUN, INRA, The Research Unit of the Thorax Institute, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, F-44000 Nantes, France
| | - Julio Sergio Marchini
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto 14000-000, São Paulo, Brazil
| |
Collapse
|
20
|
Jiang H, Li XS, Yang Y, Qi RX. Plasma lipidomics profiling in predicting the chemo-immunotherapy response in advanced non-small cell lung cancer. Front Oncol 2024; 14:1348164. [PMID: 39040440 PMCID: PMC11260645 DOI: 10.3389/fonc.2024.1348164] [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: 12/01/2023] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Background Advanced non-small cell lung cancer (NSCLC) presents significant treatment challenges, with chemo-immunotherapy emerging as a promising approach. This study explores the potential of lipidomic biomarkers to predict responses to chemo-immunotherapy in advanced non-small cell lung cancer (NSCLC) patients. Methods A prospective analysis was conducted on 68 NSCLC patients undergoing chemo-immunotherapy, divided into disease control (DC) and progressive disease (PD) groups based on treatment response. Pre-treatment serum samples were subjected to lipidomic profiling using liquid chromatography-mass spectrometry (LC-MS). Key predictive lipids (biomarkers) were identified through projection to latent structures discriminant analysis. A biomarker combined model and a clinical combined model were developed to enhance the prediction accuracy. The predictive performances of the clinical combined model in different histological subtypes were also performed. Results Six lipids were identified as the key lipids. The expression levels of PC(16:0/18:2), PC(16:0/18:1), PC(16:0/18:0), CE(20:1), and PC(14:0/18:1) were significantly up-regulated. While the expression level of TAG56:7-FA18:2 was significantly down-regulated. The biomarker combined model demonstrated a receiver operating characteristic (ROC) curve of 0.85 (95% CI: 0.75-0.95) in differentiating the PD from the DC. The clinical combined model exhibited an AUC of 0.87 (95% CI: 0.79-0.96) in differentiating the PD from the DC. The clinical combined model demonstrated good discriminability in DC and PD patients in different histological subtypes with the AUC of 0.78 (95% CI: 0.62-0.96), 0.79 (95% CI: 0.64-0.94), and 0.86 (95% CI: 0.52-1.00) in squamous cell carcinoma, large cell carcinoma, and adenocarcinoma subtype, respectively. Pathway analysis revealed the metabolisms of linoleic acid, alpha-linolenic acid, glycerolipid, arachidonic acid, glycerophospholipid, and steroid were implicated in the chemo-immunotherapy response in advanced NSCLC. Conclusion Lipidomic profiling presents a highly accurate method for predicting responses to chemo-immunotherapy in patients with advanced NSCLC, offering a potential avenue for personalized treatment strategies.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Ultrasound, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xu-Shuo Li
- Department of Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ying Yang
- Department of Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Rui-Xue Qi
- Department of Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Guo X, Ou T, Yang X, Song Q, Zhu L, Mi S, Zhang J, Zhang Y, Chen W, Guo J. Untargeted metabolomics based on ultra-high performance liquid chromatography-mass spectrometry/MS reveals the lipid-lowering mechanism of taurine in hyperlipidemia mice. Front Nutr 2024; 11:1367589. [PMID: 38706565 PMCID: PMC11066166 DOI: 10.3389/fnut.2024.1367589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Taurine has a prominent lipid-lowering effect on hyperlipidemia. However, a comprehensive analysis of the effects of taurine on endogenous metabolites in hyperlipidemia has not been documented. This study aimed to explore the impact of taurine on multiple metabolites associated with hyperlipidemia. Methods The hyperlipidemic mouse model was induced by high-fat diet (HFD). Taurine was administered via oral gavage at doses of 700 mg/kg/day for 14 weeks. Evaluation of body weight, serum lipid levels, and histopathology of the liver and adipose tissue was performed to confirm the lipid-lowering effect of taurine. Ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS)-based metabonomics analyses of serum, urine, feces, and liver, coupled with multivariate data analysis, were conducted to assess changes in the endogenous metabolites. Results and discussion Biochemical and histological examinations demonstrated that taurine administration prevented weight gain and dyslipidemia, and alleviated lipid deposition in the liver and adipose tissue in hyperlipidemic mice. A total of 76 differential metabolites were identified by UPLC-MS-based metabolomics approach, mainly involving BAs, GPs, SMs, DGs, TGs, PUFAs and amino acids. Taurine was found to partially prevent HFDinduced abnormalities in the aforementioned metabolites. Using KEGG database and MetaboAnalyst software, it was determined that taurine effectively alleviates metabolic abnormalities caused by HFD, including fatty acid metabolism, sphingolipid metabolism, glycerophospholipid metabolism, diacylglycerol metabolism, amino acid metabolism, bile acid and taurine metabolism, taurine and hypotaurine metabolism. Moreover, DGs, GPs and SMs, and taurine itself may serve as active metabolites in facilitating various anti-hyperlipidemia signal pathways associated with taurine. This study provides new evidence for taurine to prevent hyperlipidemia.
Collapse
Affiliation(s)
- Xinzhe Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Tong Ou
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Xinyu Yang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qi Song
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Lin Zhu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Shengquan Mi
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Jing Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yanzhen Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Wen Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Junxia Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| |
Collapse
|
22
|
Morgan PK, Pernes G, Huynh K, Giles C, Paul S, Smith AAT, Mellett NA, Liang A, van Buuren-Milne T, Veiga CB, Collins TJC, Xu Y, Lee MKS, De Silva TM, Meikle PJ, Lancaster GI, Murphy AJ. A lipid atlas of human and mouse immune cells provides insights into ferroptosis susceptibility. Nat Cell Biol 2024; 26:645-659. [PMID: 38589531 DOI: 10.1038/s41556-024-01377-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 02/19/2024] [Indexed: 04/10/2024]
Abstract
The cellular lipidome comprises thousands of unique lipid species. Here, using mass spectrometry-based targeted lipidomics, we characterize the lipid landscape of human and mouse immune cells ( www.cellularlipidatlas.com ). Using this resource, we show that immune cells have unique lipidomic signatures and that processes such as activation, maturation and development impact immune cell lipid composition. To demonstrate the potential of this resource to provide insights into immune cell biology, we determine how a cell-specific lipid trait-differences in the abundance of polyunsaturated fatty acid-containing glycerophospholipids (PUFA-PLs)-influences immune cell biology. First, we show that differences in PUFA-PL content underpin the differential susceptibility of immune cells to ferroptosis. Second, we show that low PUFA-PL content promotes resistance to ferroptosis in activated neutrophils. In summary, we show that the lipid landscape is a defining feature of immune cell identity and that cell-specific lipid phenotypes underpin aspects of immune cell physiology.
Collapse
Affiliation(s)
- Pooranee K Morgan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Gerard Pernes
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Sudip Paul
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Amy Liang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Thomas J C Collins
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Yangsong Xu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Man K S Lee
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - T Michael De Silva
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia.
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
23
|
Wohlwend M, Laurila PP, Goeminne LJE, Lima T, Daskalaki I, Li X, von Alvensleben G, Crisol B, Mangione R, Gallart-Ayala H, Lalou A, Burri O, Butler S, Morris J, Turner N, Ivanisevic J, Auwerx J. Inhibition of CERS1 in skeletal muscle exacerbates age-related muscle dysfunction. eLife 2024; 12:RP90522. [PMID: 38506902 PMCID: PMC10954306 DOI: 10.7554/elife.90522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Age-related muscle wasting and dysfunction render the elderly population vulnerable and incapacitated, while underlying mechanisms are poorly understood. Here, we implicate the CERS1 enzyme of the de novo sphingolipid synthesis pathway in the pathogenesis of age-related skeletal muscle impairment. In humans, CERS1 abundance declines with aging in skeletal muscle cells and, correlates with biological pathways involved in muscle function and myogenesis. Furthermore, CERS1 is upregulated during myogenic differentiation. Pharmacological or genetic inhibition of CERS1 in aged mice blunts myogenesis and deteriorates aged skeletal muscle mass and function, which is associated with the occurrence of morphological features typical of inflammation and fibrosis. Ablation of the CERS1 orthologue lagr-1 in Caenorhabditis elegans similarly exacerbates the age-associated decline in muscle function and integrity. We discover genetic variants reducing CERS1 expression in human skeletal muscle and Mendelian randomization analysis in the UK biobank cohort shows that these variants reduce muscle grip strength and overall health. In summary, our findings link age-related impairments in muscle function to a reduction in CERS1, thereby underlining the importance of the sphingolipid biosynthesis pathway in age-related muscle homeostasis.
Collapse
Affiliation(s)
- Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Ludger JE Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Tanes Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Ioanna Daskalaki
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Giacomo von Alvensleben
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Renata Mangione
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL)LausanneSwitzerland
| | - Amélia Lalou
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Olivier Burri
- Bioimaging and optics platform, École polytechnique fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Stephen Butler
- School of Chemistry, University of New South Wales SydneySydneyAustralia
| | - Jonathan Morris
- School of Chemistry, University of New South Wales SydneySydneyAustralia
| | - Nigel Turner
- Cellular Bioenergetics Laboratory, Victor Chang Cardiac Research InstituteDarlinghurstAustralia
- School of Biomedical Sciences, University of New South Wales SydneySydneyAustralia
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL)LausanneSwitzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| |
Collapse
|
24
|
Dakic A, Wu J, Wang T, Huynh K, Mellett N, Duong T, Beyene HB, Magliano DJ, Shaw JE, Carrington MJ, Inouye M, Yang JY, Figtree GA, Curran JE, Blangero J, Simes J, Giles C, Meikle PJ. Imputation of plasma lipid species to facilitate integration of lipidomic datasets. Nat Commun 2024; 15:1540. [PMID: 38378775 PMCID: PMC10879118 DOI: 10.1038/s41467-024-45838-3] [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: 04/25/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
Recent advancements in plasma lipidomic profiling methodology have significantly increased specificity and accuracy of lipid measurements. This evolution, driven by improved chromatographic and mass spectrometric resolution of newer platforms, has made it challenging to align datasets created at different times, or on different platforms. Here we present a framework for harmonising such plasma lipidomic datasets with different levels of granularity in their lipid measurements. Our method utilises elastic-net prediction models, constructed from high-resolution lipidomics reference datasets, to predict unmeasured lipid species in lower-resolution studies. The approach involves (1) constructing composite lipid measures in the reference dataset that map to less resolved lipids in the target dataset, (2) addressing discrepancies between aligned lipid species, (3) generating prediction models, (4) assessing their transferability into the targe dataset, and (5) evaluating their prediction accuracy. To demonstrate our approach, we used the AusDiab population-based cohort (747 lipid species) as the reference to impute unmeasured lipid species into the LIPID study (342 lipid species). Furthermore, we compared measured and imputed lipids in terms of parameter estimation and predictive performance, and validated imputations in an independent study. Our method for harmonising plasma lipidomic datasets will facilitate model validation and data integration efforts.
Collapse
Affiliation(s)
- Aleksandar Dakic
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Jingqin Wu
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, 3086, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, VIC, 3010, Australia
| | - Natalie Mellett
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, 3086, Australia
| | | | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Melinda J Carrington
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, VIC, 3010, Australia
| | - Michael Inouye
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Jean Y Yang
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, The University of Sydney, St Leonards, NSW, 2065, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine at University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine at University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - John Simes
- National Health and Medical Research Council of Australia (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, 3086, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, VIC, 3010, Australia.
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, 3086, Australia.
- Baker Department of Cardiometabolic Health, The University of Melbourne, VIC, 3010, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
25
|
Saprina TV, Bashirova AS, Ivanov VV, Pekov SI, Popov IA, Bashirov SR, Vasilyeva EA, Pavlenko OA, Krinitskii DV, Chen M. Lipidomic markers of obesity and their dynamics after bariatric surgery. BULLETIN OF SIBERIAN MEDICINE 2024; 22:174-187. [DOI: 10.20538/1682-0363-2023-4-174-187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Obesity is considered as a chronic progressive disease, heterogeneous in its etiology and clinical manifestations, and characterized by excess in body fat mass and its deposition in the body. The term “morbid obesity” refers to excessive deposition of adipose tissue with a body mass index (BMI) ≥40 kg / m2 or with a BMI ≥ 35 kg / m2 in the presence of serious complications associated with obesity. Along with obesity, the frequency of type 2 diabetes mellitus and cardiovascular diseases closely associated with it has increased. It results from the progression of metabolic disorders, including insulin resistance, which is inextricably linked with the accumulation of visceral fat and plays a key role in the pathogenesis of obesity-related diseases.The study of lipidomic signatures in obesity and associated conditions is a promising branch of fundamental medicine, which makes it possible to significantly and at a new conceptual level stratify a cohort of obese patients into various phenotypes, including a metabolically healthy and metabolically unhealthy obesity phenotypes. Dynamic changes in the lipidome both in the context of diet, drug treatment, and after various bariatric surgeries are of great interest for developing personalized strategies for the treatment of this disease. Currently available studies and their results suggest that we are only at the very start of studying this promising biomedical field.
Collapse
Affiliation(s)
| | | | | | - S. I. Pekov
- Siberian State Medical University;
Skolkovo Institute of Science and Technology;
Moscow Institute of Physics and Technology
| | - I. A. Popov
- Siberian State Medical University;
Moscow Institute of Physics and Technology
| | | | | | | | | | - M. Chen
- Siberian State Medical University
| |
Collapse
|
26
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
27
|
Barosova R, Baranovicova E, Hanusrichterova J, Mokra D. Metabolomics in Animal Models of Bronchial Asthma and Its Translational Importance for Clinics. Int J Mol Sci 2023; 25:459. [PMID: 38203630 PMCID: PMC10779398 DOI: 10.3390/ijms25010459] [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: 11/15/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Bronchial asthma is an extremely heterogenous chronic respiratory disorder with several distinct endotypes and phenotypes. These subtypes differ not only in the pathophysiological changes and/or clinical features but also in their response to the treatment. Therefore, precise diagnostics represent a fundamental condition for effective therapy. In the diagnostic process, metabolomic approaches have been increasingly used, providing detailed information on the metabolic alterations associated with human asthma. Further information is brought by metabolomic analysis of samples obtained from animal models. This article summarizes the current knowledge on metabolomic changes in human and animal studies of asthma and reveals that alterations in lipid metabolism, amino acid metabolism, purine metabolism, glycolysis and the tricarboxylic acid cycle found in the animal studies resemble, to a large extent, the changes found in human patients with asthma. The findings indicate that, despite the limitations of animal modeling in asthma, pre-clinical testing and metabolomic analysis of animal samples may, together with metabolomic analysis of human samples, contribute to a novel way of personalized treatment of asthma patients.
Collapse
Affiliation(s)
- Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Eva Baranovicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
28
|
Jeon S, Scorletti E, Dempsey J, Buyco D, Lin C, Saiman Y, Bayen S, Harkin J, Martin J, Hooks R, Ogretmen B, Argemi J, Melo L, Bataller R, Carr RM. Ceramide synthase 6 (CerS6) is upregulated in alcohol-associated liver disease and exhibits sex-based differences in the regulation of energy homeostasis and lipid droplet accumulation. Mol Metab 2023; 78:101804. [PMID: 37714377 PMCID: PMC10561121 DOI: 10.1016/j.molmet.2023.101804] [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: 05/26/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE Alcohol-associated liver disease (ALD) is the leading cause of liver-related mortality worldwide. Current strategies to manage ALD focus largely on advanced stage disease, however, metabolic changes such as glucose intolerance are apparent at the earliest stage of alcoholic steatosis and increase the risk of disease progression. Ceramides impair insulin signaling and accumulate in ALD, and metabolic pathways involving ceramide synthase 6 (CerS6) are perturbed in ALD during hepatic steatosis. In this study, we aimed to investigate the role of CerS6 in ALD development and the relevance of CerS6 to human ALD. METHODS C57BL/6 WT and CerS6 KO mice of both sexes were fed either a Lieber-DeCarli control (CON) or 15% ethanol (EtOH) diet for six weeks. In vivo metabolic tests including glucose and insulin tolerance tests (GTT and ITT) and energy expenditure were performed. The mice were euthanized, and serum and liver lipids and liver histology were examined. For in vitro studies, CerS6 was deleted in human hepatocytes, VL17A and cells were incubated with EtOH and/or C16:0-ceramides. RNAseq analysis was performed in livers from mice and human patients with different stages of ALD and diseased controls. RESULTS After six weeks on an EtOH diet, CerS6 KO mice had reduced body weight, food intake, and %fat mass compared to WT mice. Energy expenditure increased in both male and female KO mice, however, was only statistically significant in male mice. In response to EtOH, WT mice developed mild hepatic steatosis, while steatosis was ameliorated in KO mice as determined by H&E and ORO staining. KO mice showed significantly decreased long-chain ceramide species, especially C16:0-ceramides, in the serum and liver tissues compared to WT mice. CerS6 deletion decreased serum TG and NEFA only in male not female mice. CerS6 deletion improved glucose tolerance and insulin resistance in EtOH-fed mice of both sexes. RNAseq analysis revealed that 74 genes are significantly upregulated and 66 genes are downregulated by CerS6 deletion in EtOH-fed male mice, with key network pathways including TG biosynthetic process, positive regulation of lipid localization, and fat cell differentiation. Similar to RNAseq results, absence of CerS6 significantly decreased mRNA expression of lipid droplet associated proteins in EtOH-fed mice. In vitro, EtOH stimulation significantly increased PLIN2 protein expression in VL17A cells while CerS6 deletion inhibited EtOH-mediated PLIN2 upregulation. C16:0-ceramide treatment significantly increased PLIN2 protein expression compared to CON. Notably, progression of ALD in humans was associated with increased hepatic CerS6 expression. CONCLUSIONS Our findings demonstrate that CerS6 deletion improves glucose homeostasis in alcohol-fed mice and exhibits sex-based differences in the attenuation of EtOH-induced weight gain and hepatic steatosis. Additionally, we unveil that CerS6 plays a major role as a regulator of lipid droplet biogenesis in alcohol-induced intra-hepatic lipid droplet formation, identifying it as a putative target for early ALD management.
Collapse
Affiliation(s)
- Sookyoung Jeon
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA; Department of Food Science & Nutrition and the Korean Institute of Nutrition, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
| | - Eleonora Scorletti
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph Dempsey
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - Delfin Buyco
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsea Lin
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yedidya Saiman
- Department of Medicine, Section of Hepatology, Lewis Katz School of Medicine Temple University, Philadelphia, PA, USA
| | - Susovon Bayen
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - Julia Harkin
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - Jasmin Martin
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Royce Hooks
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Josepmaria Argemi
- Center for Liver Diseases, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Luma Melo
- Center for Liver Diseases, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ramon Bataller
- Center for Liver Diseases, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rotonya M Carr
- Division of Gastroenterology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
29
|
Lee CH, Wang CY, Kao HL, Wu WK, Kuo CH. Differentiation of Alkyl- and Plasmenyl-phosphatidylcholine by Endogenous Sphingomyelin RT-XLOGP3 Regression for Coronary Artery Disease Plasma Lipidomics Analysis. Anal Chem 2023; 95:16902-16910. [PMID: 37931321 DOI: 10.1021/acs.analchem.3c02693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Accurate identification between alkyl- and plasmenyl-phosphatidylcholine (PC(O-) and PC(P-)) isomers is a major analytical challenge in lipidomics studies due to a lack of structure-specific ions in conventional tandem mass spectrometry (MS/MS) methods and the absence of universal retention time (RT) references. Given the importance of PC(O-) and PC(P-), an easy-to-apply method for current research is urgently needed. In this study, we present a quadratic RT-XLOGP3SM regression model that uses endogenous sphingomyelin (SM) species in blood samples as retention time (RT) indicators to predict the RTs of PC(O-) and PC(P-) species by coupling their calculated partition coefficients based on XLOGP3. The prediction results were obtained with a root-mean-square error (RMSE) of 0.12 min (1.3%) for the RRHD (rapid resolution high definition) nonlinear LC condition. A lipidomic analysis with RT-XLOGP3SM regression was used to study lipid regulation in coronary artery disease (CAD) outpatient plasma samples, and we found that the types of exhibited regulation were highly dependent on the lipid subclasses in comparison to the healthy control group. In conclusion, given that the quadratic RT-XLOGP3SM regression model predicts the RTs of PC species based on the relative value of XLOGP3 and the RTs of endogenous SM species, it can be expected that most of the C18-based lipidomics analyses could apply this method to increase the identification ability of the PC(O-) and PC(P-) subclasses and to improve the understanding of their physiological functions.
Collapse
Affiliation(s)
- Ching-Hua Lee
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 10050, Taiwan
| | - Chin-Yi Wang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 10050, Taiwan
| | - Hsien-Li Kao
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 10048, Taiwan
| | - Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei 10048, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 10050, Taiwan
- Department of Pharmacy, National Taiwan University Hospital, Taipei 10048, Taiwan
| |
Collapse
|
30
|
Tabassum R, Widén E, Ripatti S. Effect of biological sex on human circulating lipidome: An overview of the literature. Atherosclerosis 2023; 384:117274. [PMID: 37743161 DOI: 10.1016/j.atherosclerosis.2023.117274] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/28/2023] [Accepted: 09/01/2023] [Indexed: 09/26/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide for both men and women, but their prevalence and burden show marked sex differences. The existing knowledge gaps in research, prevention, and treatment for women emphasize the need for understanding the biological mechanisms contributing to the sex differences in CVD. Sex differences in the plasma lipids that are well-known risk factors and predictors of CVD events have been recognized and are believed to contribute to the known disparities in CVD manifestations in men and women. However, the current understanding of sex differences in lipids has mainly come from the studies on routinely measured standard lipids- low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), total triglycerides, and total cholesterol, which have been the mainstay of the lipid profiling. Sex differences in individual lipid species, collectively called the lipidome, have until recently been less explored due to the technological challenges and analytic costs. With the technological advancements in the last decade and growing interest in understanding mechanisms of sexual dimorphism in metabolic disorders, many investigators utilized metabolomics and lipidomics based platforms to examine the effect of biological sex on detailed lipidomic profiles and individual lipid species. This review presents an overview of the research on sex differences in the concentrations of circulating lipid species, focusing on findings from the metabolome- and lipidome-wide studies. We also discuss the potential contribution of genetic factors including sex chromosomes and sex-specific physiological factors such as menopause and sex hormones to the sex differences in lipidomic profiles.
Collapse
Affiliation(s)
- Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
31
|
Li Y, Chaurasia B, Rahman MM, Kaddai V, Maschek JA, Berg JA, Wilkerson JL, Mahmassani ZS, Cox J, Wei P, Meikle PJ, Atkinson D, Wang L, Poss AM, Playdon MC, Tippetts TS, Mousa EM, Nittayaboon K, Anandh Babu PV, Drummond MJ, Clevers H, Shayman JA, Hirabayashi Y, Holland WL, Rutter J, Edgar BA, Summers SA. Ceramides Increase Fatty Acid Utilization in Intestinal Progenitors to Enhance Stemness and Increase Tumor Risk. Gastroenterology 2023; 165:1136-1150. [PMID: 37541526 PMCID: PMC10592225 DOI: 10.1053/j.gastro.2023.07.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND & AIMS Cancers of the alimentary tract, including esophageal adenocarcinomas, colorectal cancers, and cancers of the gastric cardia, are common comorbidities of obesity. Prolonged, excessive delivery of macronutrients to the cells lining the gut can increase one's risk for these cancers by inducing imbalances in the rate of intestinal stem cell proliferation vs differentiation, which can produce polyps and other aberrant growths. We investigated whether ceramides, which are sphingolipids that serve as a signal of nutritional excess, alter stem cell behaviors to influence cancer risk. METHODS We profiled sphingolipids and sphingolipid-synthesizing enzymes in human adenomas and tumors. Thereafter, we manipulated expression of sphingolipid-producing enzymes, including serine palmitoyltransferase (SPT), in intestinal progenitors of mice, cultured organoids, and Drosophila to discern whether sphingolipids altered stem cell proliferation and metabolism. RESULTS SPT, which diverts dietary fatty acids and amino acids into the biosynthetic pathway that produces ceramides and other sphingolipids, is a critical modulator of intestinal stem cell homeostasis. SPT and other enzymes in the sphingolipid biosynthesis pathway are up-regulated in human intestinal adenomas. They produce ceramides, which serve as prostemness signals that stimulate peroxisome-proliferator activated receptor-α and induce fatty acid binding protein-1. These actions lead to increased lipid utilization and enhanced proliferation of intestinal progenitors. CONCLUSIONS Ceramides serve as critical links between dietary macronutrients, epithelial regeneration, and cancer risk.
Collapse
Affiliation(s)
- Ying Li
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Bhagirath Chaurasia
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah; Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa.
| | - M Mahidur Rahman
- Huntsman Cancer Institute and Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| | - Vincent Kaddai
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - J Alan Maschek
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Jordan A Berg
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
| | - Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah
| | - James Cox
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
| | - Peng Wei
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Donald Atkinson
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Liping Wang
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Annelise M Poss
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Mary C Playdon
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Trevor S Tippetts
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Esraa M Mousa
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah; Faculty of Science, Tanta University, Tanta, Egypt
| | - Kesara Nittayaboon
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah; Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands; Oncode Institute, Utrecht, The Netherlands; Princess Maxima Center (PMC) for Pediatric Oncology, Utrecht, The Netherlands
| | - James A Shayman
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yoshio Hirabayashi
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako-shi, Saitama Japan
| | - William L Holland
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, Utah; Howard Hughes Medical Institute, Salt Lake City, Utah
| | - Bruce A Edgar
- Huntsman Cancer Institute and Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
32
|
George AD, Paul S, Wang T, Huynh K, Giles C, Mellett N, Duong T, Nguyen A, Geddes D, Mansell T, Saffery R, Vuillermin P, Ponsonby AL, Burgner D, Burugupalli S, Meikle PJ. Defining the lipid profiles of human milk, infant formula, and animal milk: implications for infant feeding. Front Nutr 2023; 10:1227340. [PMID: 37712002 PMCID: PMC10499237 DOI: 10.3389/fnut.2023.1227340] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/15/2023] [Indexed: 09/16/2023] Open
Abstract
Background Breastfed infants have lower disease risk compared to formula-fed infants, however, the mechanisms behind this protection are unknown. Human milk has a complex lipidome which may have many critical roles in health and disease risk. However, human milk lipidomics is challenging, and research is still required to fully understand the lipidome and to interpret and translate findings. This study aimed to address key human milk lipidome knowledge gaps and discuss possible implications for early life health. Methods Human milk samples from two birth cohorts, the Barwon Infant Study (n = 312) and University of Western Australia birth cohort (n = 342), were analysed using four liquid chromatography-mass spectrometry (LC-MS) methods (lipidome, triacylglycerol, total fatty acid, alkylglycerol). Bovine, goat, and soy-based infant formula, and bovine and goat milk were analysed for comparison. Composition was explored as concentrations, relative abundance, and infant lipid intake. Statistical analyses included principal component analysis, mixed effects modelling, and correlation, with false discovery rate correction, to explore human milk lipidome longitudinal trends and inter and intra-individual variation, differences between sample types, lipid intakes, and correlations between infant plasma and human milk lipids. Results Lipidomics analysis identified 979 lipids. The human milk lipidome was distinct from that of infant formula and animal milk. Ether lipids were of particular interest, as they were significantly higher, in concentration and relative abundance, in human milk than in formula and animal milk, if present in the latter samples at all. Many ether lipids were highest in colostrum, and some changed significantly through lactation. Significant correlations were identified between human milk and infant circulating lipids (40% of which were ether lipids), and specific ether lipid intake by exclusively breastfed infants was 200-fold higher than that of an exclusively formula-fed infant. Conclusion There are marked differences between the lipidomes of human milk, infant formula, and animal milk, with notable distinctions between ether lipids that are reflected in the infant plasma lipidome. These findings have potential implications for early life health, and may reveal why breast and formula-fed infants are not afforded the same protections. Comprehensive lipidomics studies with outcomes are required to understand the impacts on infant health and tailor translation.
Collapse
Affiliation(s)
- Alexandra D. George
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Sudip Paul
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Tingting Wang
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Corey Giles
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Natalie Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Thy Duong
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Anh Nguyen
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Toby Mansell
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Richard Saffery
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Peter Vuillermin
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- School of Medicine, Deakin University, Melbourne, VIC, Australia
- Child Health Research Unit, Barwon Health, Geelong, VIC, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - David Burgner
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Satvika Burugupalli
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Peter J. Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| |
Collapse
|
33
|
Bartho LA, Keenan E, Walker SP, MacDonald TM, Nijagal B, Tong S, Kaitu'u-Lino TJ. Plasma lipids are dysregulated preceding diagnosis of preeclampsia or delivery of a growth restricted infant. EBioMedicine 2023; 94:104704. [PMID: 37421807 PMCID: PMC10344703 DOI: 10.1016/j.ebiom.2023.104704] [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: 03/13/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Lipids serve as multifunctional metabolites that have important implications for the pregnant mother and developing fetus. Abnormalities in lipids have emerged as potential risk factors for pregnancy diseases, such as preeclampsia and fetal growth restriction. The aim of this study was to assess the potential of lipid metabolites for detection of late-onset preeclampsia and fetal growth restriction. METHODS We used a case-cohort of 144 maternal plasma samples at 36 weeks' gestation from patients before the diagnosis of late-onset preeclampsia (n = 22), delivery of a fetal growth restricted infant (n = 55, defined as <5th birthweight centile), gestation-matched controls (n = 72). We performed liquid chromatography-tandem mass spectrometry (LC-QQQ) -based targeted lipidomics to identify 421 lipids, and fitted logistic regression models for each lipid, correcting for maternal age, BMI, smoking, and gestational diabetes. FINDINGS Phosphatidylinositol 32:1 (AUC = 0.81) and cholesterol ester 17:1 (AUC = 0.71) best predicted the risk of developing preeclampsia or delivering a fetal growth restricted infant, respectively. Five times repeated five-fold cross validation demonstrated the lipids alone did not out-perform existing protein biomarkers, soluble tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF) for the prediction of preeclampsia or fetal growth restriction. However, lipids combined with sFlt-1 and PlGF measurements improved disease prediction. INTERPRETATION This study successfully identified 421 lipids in maternal plasma collected at 36 weeks' gestation from participants who later developed preeclampsia or delivered a fetal growth restricted infant. Our results suggest the predictive capacity of lipid measurements for gestational disorders holds the potential to improve non-invasive assessment of maternal and fetal health. FUNDING This study was funded by a grant from National Health and Medical Research Council.
Collapse
Affiliation(s)
- Lucy A Bartho
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia.
| | - Emerson Keenan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Susan P Walker
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Teresa M MacDonald
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Brunda Nijagal
- Metabolomics Australia, The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| |
Collapse
|
34
|
Barrachina MN, Pernes G, Becker IC, Allaeys I, Hirsch TI, Groeneveld DJ, Khan AO, Freire D, Guo K, Carminita E, Morgan PK, Collins TJC, Mellett NA, Wei Z, Almazni I, Italiano JE, Luyendyk J, Meikle PJ, Puder M, Morgan NV, Boilard E, Murphy AJ, Machlus KR. Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36. NATURE CARDIOVASCULAR RESEARCH 2023; 2:746-763. [PMID: 39195958 PMCID: PMC11909960 DOI: 10.1038/s44161-023-00305-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/21/2023] [Indexed: 08/29/2024]
Abstract
Lipids contribute to hematopoiesis and membrane properties and dynamics; however, little is known about the role of lipids in megakaryopoiesis. Here we show that megakaryocyte progenitors, megakaryocytes and platelets present a unique lipidome progressively enriched in polyunsaturated fatty acid (PUFA)-containing phospholipids. In vitro, inhibition of both exogenous fatty acid functionalization and uptake as well as de novo lipogenesis impaired megakaryocyte differentiation and proplatelet production. In vivo, mice on a high saturated fatty acid diet had significantly lower platelet counts, which was prevented by eating a PUFA-enriched diet. Fatty acid uptake was largely dependent on CD36, and its deletion in mice resulted in low platelets. Moreover, patients with a CD36 loss-of-function mutation exhibited thrombocytopenia and increased bleeding. Our results suggest that fatty acid uptake and regulation is essential for megakaryocyte maturation and platelet production and that changes in dietary fatty acids may be a viable target to modulate platelet counts.
Collapse
Affiliation(s)
- Maria N Barrachina
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Gerard Pernes
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Isabelle C Becker
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Isabelle Allaeys
- Centre de Recherche du CHU de Québec-Université Laval and Centre de Recherche ARThrite, Québec, QC, Canada
| | - Thomas I Hirsch
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Dafna J Groeneveld
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Daniela Freire
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
| | - Karen Guo
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
| | - Estelle Carminita
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Pooranee K Morgan
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Thomas J C Collins
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Natalie A Mellett
- Metabolomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Zimu Wei
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Ibrahim Almazni
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - James Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Peter J Meikle
- Metabolomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eric Boilard
- Centre de Recherche du CHU de Québec-Université Laval and Centre de Recherche ARThrite, Québec, QC, Canada
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
35
|
Hildebrand F, Schoeny H, Rampler E, Koellensperger G. Scrutinizing different ionization responses of polar lipids in a reversed-phase gradient by implementing a counter-gradient. Anal Chim Acta 2023; 1265:341274. [PMID: 37230568 DOI: 10.1016/j.aca.2023.341274] [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: 02/14/2023] [Revised: 03/30/2023] [Accepted: 04/23/2023] [Indexed: 05/27/2023]
Abstract
Lipidomics studies strive for a comprehensive identification and quantification of lipids. While reversed phase (RP) liquid chromatography (LC) coupled to high resolution mass spectrometry (MS) offers unrivalled selectivity and thus is the preferred method for lipid identification, accurate lipid quantification remains challenging. The widely adopted one-point lipid class specific quantification (one internal standard per lipid class) suffers from the fact that ionization of internal standard and target lipid occurs under different solvent composition as a consequence of chromatographic separation. To address this issue, we established a dual flow injection and chromatography setup that allows to control solvent conditions during ionization enabling isocratic ionization while running a RP gradient through the use of a counter-gradient. Using this dual LC pump platform, we investigated the impact of solvent conditions within a RP gradient on ionization response and arising quantification biases. Our results confirmed that changing solvent composition significantly influences ionization response. Quantification of human plasma (SRM 1950) lipids under gradient and isocratic ionization conditions further confirmed these findings as significant differences between the two conditions were found for the majority of lipids. While the quantity of sphingomyelins with >40 C atoms was consistently overestimated under gradient ionization, isocratic ionization improved their recovery compared to consensus values. However, the limitation of consensus values was demonstrated as overall only small changes in z-score were observed because of high uncertainties of the consensus values. Furthermore, we observed a trueness bias between gradient and isocratic ionization when quantifying a panel of lipid species standards which is highly dependent on lipid class and ionization mode. Uncertainty calculations under consideration of the trueness bias as RP gradient uncertainty revealed that especially ceramides with >40 C atoms had a high bias leading to total combined uncertainties of up to 54%. The assumption of isocratic ionization significantly decreases total measurement uncertainty and highlights the importance of studying the trueness bias introduced by a RP gradient to reduce quantification uncertainty.
Collapse
Affiliation(s)
- Felina Hildebrand
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria; Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
| | - Harald Schoeny
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
| | - Evelyn Rampler
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Althanstr. 14, 1090, Vienna, Austria.
| |
Collapse
|
36
|
Polzin A, Dannenberg L, Benkhoff M, Barcik M, Helten C, Mourikis P, Ahlbrecht S, Wildeis L, Ziese J, Zikeli D, Metzen D, Hu H, Baensch L, Schröder NH, Keul P, Weske S, Wollnitzke P, Duse D, Saffak S, Cramer M, Bönner F, Müller T, Gräler MH, Zeus T, Kelm M, Levkau B. Revealing concealed cardioprotection by platelet Mfsd2b-released S1P in human and murine myocardial infarction. Nat Commun 2023; 14:2404. [PMID: 37100836 PMCID: PMC10133218 DOI: 10.1038/s41467-023-38069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
Antiplatelet medication is standard of care in acute myocardial infarction (AMI). However, it may have obscured beneficial properties of the activated platelet secretome. We identify platelets as major source of a sphingosine-1-phosphate (S1P) burst during AMI, and find its magnitude to favorably associate with cardiovascular mortality and infarct size in STEMI patients over 12 months. Experimentally, administration of supernatant from activated platelets reduces infarct size in murine AMI, which is blunted in platelets deficient for S1P export (Mfsd2b) or production (Sphk1) and in mice deficient for cardiomyocyte S1P receptor 1 (S1P1). Our study reveals an exploitable therapeutic window in antiplatelet therapy in AMI as the GPIIb/IIIa antagonist tirofiban preserves S1P release and cardioprotection, whereas the P2Y12 antagonist cangrelor does not. Here, we report that platelet-mediated intrinsic cardioprotection is an exciting therapeutic paradigm reaching beyond AMI, the benefits of which may need to be considered in all antiplatelet therapies.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maike Barcik
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Carolin Helten
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Laura Wildeis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Justus Ziese
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dorothee Zikeli
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hao Hu
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leonard Baensch
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dragos Duse
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Süreyya Saffak
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mareike Cramer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Florian Bönner
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Tina Müller
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
37
|
Ding D, He X, Agarry IE, Wang Y, Zhou F, Li Y, Kan J, Cai T, Chen K. Profile of Human Milk Phospholipids at Different Lactation Stages with UPLC/Q-TOF-MS: Characterization, Distribution, and Differences. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6326-6337. [PMID: 37040528 DOI: 10.1021/acs.jafc.2c07512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Human milk phospholipids are important for the regular growth and development of infants. Ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF-MS) was employed to qualitatively and quantitatively analyze 277 phospholipid molecular species in 112 human milk samples to obtain a detailed profile of human milk phospholipids along the lactation stage. MS/MS fragmentation patterns of sphingomyelin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, and phosphatidylserine were characterized in detail. Phosphatidylcholine is the most dominant group, followed by sphingomyelin. PC(18:0/18:2), SM(d18:1/24:1), PE(18:0/18:0), PS(18:0/20:4), and PI(18:0/18:2) showed the highest average concentration among all of the phosphatidylcholine, sphingomyelin, phosphatidylethanolamine, phosphatidylserine, and phosphatidylinositol molecular species, respectively. The fatty acids attached to the phospholipid molecules were mainly palmitic, stearic, oleic, and linoleic acids, and the plasmalogens decreased along the lactation stage. The increase of sphingomyelins and phosphatidylethanolamines and the decrease of phosphatidylcholines are the key changes from colostrum to transitional milk; the increase of lysophosphatidylcholines and lysophosphatidylethanolamines and the continuous decrease of phosphatidylcholines are the vital changes from transitional milk to mature milk.
Collapse
Affiliation(s)
- Desheng Ding
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Xiaoling He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P. R. China
| | - Israel Emiezi Agarry
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Yuankai Wang
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Fenglan Zhou
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Yunchang Li
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jianquan Kan
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Tian Cai
- School of Chemistry and Chemical Engineering, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
| | - Kewei Chen
- College of Food Science, Southwest University, No. 2, Tiansheng Road, Beibei, Chongqing 400715, P. R. China
- Chinese-Hungarian Cooperative Research Centre for Food Science, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Specialty Food Co-built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| |
Collapse
|
38
|
Fang H, Cao Y, Zhang J, Wang X, Li M, Hong Z, Wu Z, Fang M. Lipidome remodeling activities of DPA-EA in palmitic acid-stimulated HepG2 cells and the in vivo anti-obesity effect of the DPA-EA and DHA-EA mixture prepared from algae oil. Front Pharmacol 2023; 14:1146276. [PMID: 37063272 PMCID: PMC10090563 DOI: 10.3389/fphar.2023.1146276] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Background: The nuclear receptor Nur77 has been demonstrated to play a vital role in the inflammatory response and cellular metabolisms, and its ligands exhibit efficacy in the treatment of inflammation-related diseases (e.g., improving mouse acute lung injury (ALI) and obesity. Recently, ω-3 polyunsaturated fatty acid-ethanolamine derivatives (ω-3 PUFA-EAs), including DPA-EA and DHA-EA, have been reported as new Nur77-targeting anti-inflammatory agents. However, the lipid-lowering effect of ω-3 PUFA-EAs is still unknown, and lipid profile changes induced by Nur77-targeting anti-inflammatory agents also remain unclear.Objective: This study aimed to evaluate the lipid-lowering effect and the underlying mechanism of DPA-EA acting as Nur77-targeting anti-inflammatory agents. It also aimed to investigate the in vitro and in vivo lipid-lowering effects of the DPA-EA and DHA-EA mixture prepared from algae oil.Methods: The in vitro lipid-lowing effect of DPA-EA and its mixture with DHA-EA was first evaluated in palmitic acid-stimulated HepG2 Cells. To confirm the lipid-lowering effect and explore the underlying mechanism, we performed untargeted lipidomic analysis using ultra-performance liquid chromatography/triple quadrupole-time-of-flight (TOF) mass spectrometry coupled with multivariate statistical analysis, with another Nur77-targeting anti-inflammatory compound Celastrol (Cel) as a reference. Finally, we examined the anti-obesity effect of the DPA-EA and DHA-EA mixture synthesized from algae oil in a high-fat diet (HFD)-fed mice model.Results: DPA-EA significantly alleviated lipid accumulation with lower toxicity than Celastrol. Nur77-targeting compounds DPA-EA and Celastrol could simultaneously reduce 14 lipids (9 TGs, 2 PCs, 1 PA, 1 SM, and 1 LacCer) and increase 13 lipids (4 DGs, 6 LPEs, 2 PEs, and 1PC) in Pal-stimulated HepG2 cells. However, Cer lipids were more sensitive to DPA-EA, while the over-downregulation of SM lipids might be associated with the off-target toxicity of Celastrol. The mixture of DPA-EA and DHA-EA synthesized from algae oil could significantly decrease TG, TC, and LDL levels and increase HDL levels in HFD-fed mice, exerting an excellent anti-obesity effect.Conclusion: Nur77-targeting anti-inflammatory compound DAP-EA could promote the hydrolysis of PEs and TGs to ameliorate lipid accumulation. The DPA-EA and DHA-EA mixture prepared from algae oil might be a potential therapeutic agent for obesity and other inflammation-related diseases.
Collapse
Affiliation(s)
- Hua Fang
- Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Yin Cao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jianyu Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiumei Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Mengyu Li
- Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, China
| | - Zhuan Hong
- Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Zhen Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Meijuan Fang,
| |
Collapse
|
39
|
Kvasnička A, Najdekr L, Dobešová D, Piskláková B, Ivanovová E, Friedecký D. Clinical lipidomics in the era of the big data. Clin Chem Lab Med 2023; 61:587-598. [PMID: 36592414 DOI: 10.1515/cclm-2022-1105] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023]
Abstract
Lipidomics as a branch of metabolomics provides unique information on the complex lipid profile in biological materials. In clinically focused studies, hundreds of lipids together with available clinical information proved to be an effective tool in the discovery of biomarkers and understanding of pathobiochemistry. However, despite the introduction of lipidomics nearly twenty years ago, only dozens of big data studies using clinical lipidomics have been published to date. In this review, we discuss the lipidomics workflow, statistical tools, and the challenges of standartisation. The consequent summary divided into major clinical areas of cardiovascular disease, cancer, diabetes mellitus, neurodegenerative and liver diseases is demonstrating the importance of clinical lipidomics. In these publications, the potential of lipidomics for prediction, diagnosis or finding new targets for the treatment of selected diseases can be seen. The first of these results have already been implemented in clinical practice in the field of cardiovascular diseases, while in other areas we can expect the application of the results summarized in this review in the near future.
Collapse
Affiliation(s)
- Aleš Kvasnička
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Lukáš Najdekr
- Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czechia
| | - Dana Dobešová
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Barbora Piskláková
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Eliška Ivanovová
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - David Friedecký
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| |
Collapse
|
40
|
Barrachina MN, Pernes G, Becker IC, Allaeys I, Hirsch TI, Groeneveld DJ, Khan AO, Freire D, Guo K, Carminita E, Morgan PK, Collins TJ, Mellett NA, Wei Z, Almazni I, Italiano JE, Luyendyk J, Meikle PJ, Puder M, Morgan NV, Boilard E, Murphy AJ, Machlus KR. Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.12.527706. [PMID: 36798332 PMCID: PMC9934665 DOI: 10.1101/2023.02.12.527706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Lipids contribute to hematopoiesis and membrane properties and dynamics, however, little is known about the role of lipids in megakaryopoiesis. Here, a lipidomic analysis of megakaryocyte progenitors, megakaryocytes, and platelets revealed a unique lipidome progressively enriched in polyunsaturated fatty acid (PUFA)-containing phospholipids. In vitro, inhibition of both exogenous fatty acid functionalization and uptake and de novo lipogenesis impaired megakaryocyte differentiation and proplatelet production. In vivo, mice on a high saturated fatty acid diet had significantly lower platelet counts, which was prevented by eating a PUFA-enriched diet. Fatty acid uptake was largely dependent on CD36, and its deletion in mice resulted in thrombocytopenia. Moreover, patients with a CD36 loss-of-function mutation exhibited thrombocytopenia and increased bleeding. Our results suggest that fatty acid uptake and regulation is essential for megakaryocyte maturation and platelet production, and that changes in dietary fatty acids may be a novel and viable target to modulate platelet counts.
Collapse
Affiliation(s)
- Maria N Barrachina
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Gerard Pernes
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Isabelle C Becker
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Isabelle Allaeys
- Centre de Recherche du CHU de Québec-Université Laval and Centre de Recherche ARThrite, Québec, QC, G1V4G2 Canada
| | - Thomas I. Hirsch
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Dafna J Groeneveld
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Abdullah O. Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K, B15 2TT
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine and National Institute of Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, U.K. OX3 9DS
| | - Daniela Freire
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Karen Guo
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Estelle Carminita
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Pooranee K Morgan
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Thomas J Collins
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Natalie A Mellett
- Metabolomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Zimu Wei
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Ibrahim Almazni
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K, B15 2TT
| | - Joseph E. Italiano
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - James Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Peter J Meikle
- Metabolomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Puder
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| | - Neil V. Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K, B15 2TT
| | - Eric Boilard
- Centre de Recherche du CHU de Québec-Université Laval and Centre de Recherche ARThrite, Québec, QC, G1V4G2 Canada
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA, 02115 USA
- Harvard Medical School, Department of Surgery, Boston Children’s Hospital, Boston, MA, 02115 USA
| |
Collapse
|
41
|
Stepwise solid phase extraction integrated with chemical derivatization for all-in-one injection LC-MS/MS analysis of metabolome and lipidome. Anal Chim Acta 2023; 1241:340807. [PMID: 36657877 DOI: 10.1016/j.aca.2023.340807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
The metabolome and lipidome are critical components in illustrating biological processes and pathological mechanisms. Generally, two or more independent methods are required to analyze the two compound panels due to their distinct chemical properties and polarity differences. Here, a novel strategy integrating stepwise solid-phase extraction (SPE) and dansyl chemical derivatization was proposed for all-in-one injection LC-MS/MS analysis of serum metabolome and lipidome. In this workflow, a stepwise elution procedure was firstly optimized to separate the metabolome and lipidome fractions using an Ostro plate. Dansyl chemical derivatization was then applied to label amine/phenol, carboxyl, and carbonyl-containing sub-metabolomes. Our results demonstrated that the dansyl labeling could significantly improve chromatographic separation, enhance the MS response, and overcome the matrix effect of co-eluting lipids. Ultimately, an all-in-one injection LC-MS/MS method measuring 256 lipids (covering 20 subclasses) and 212 metabolites (including amino acids, bile acids, fatty acids, acylcarnitines, indole derivatives, ketones and aldehydes, nucleic acid metabolism, polyamines, etc.) was established. This method was applied to investigate the metabolic changes in cisplatin-induced nephrotoxicity in rats and the results were compared with previous untargeted metabolomics. The presented strategy could predominantly improve the analytical coverage and throughput and can be of great use in discovering reliable potential biomarkers in various applications.
Collapse
|
42
|
Kalinichenko LS, Mühle C, Jia T, Anderheiden F, Datz M, Eberle AL, Eulenburg V, Granzow J, Hofer M, Hohenschild J, Huber SE, Kämpf S, Kogias G, Lacatusu L, Lugmair C, Taku SM, Meixner D, Sembritzki NK, Praetner M, Rhein C, Sauer C, Scholz J, Ulrich F, Valenta F, Weigand E, Werner M, Tay N, Mc Veigh CJ, Haase J, Wang AL, Abdel-Hafiz L, Huston JP, Smaga I, Frankowska M, Filip M, Lourdusamy A, Kirchner P, Ekici AB, Marx LM, Suresh NP, Frischknecht R, Fejtova A, Saied EM, Arenz C, Bozec A, Wank I, Kreitz S, Hess A, Bäuerle T, Ledesma MD, Mitroi DN, Miranda AM, Oliveira TG, Lenz B, Schumann G, Kornhuber J, Müller CP. Adult alcohol drinking and emotional tone are mediated by neutral sphingomyelinase during development in males. Cereb Cortex 2023; 33:844-864. [PMID: 35296883 DOI: 10.1093/cercor/bhac106] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/03/2023] Open
Abstract
Alcohol use, abuse, and addiction, and resulting health hazards are highly sex-dependent with unknown mechanisms. Previously, strong links between the SMPD3 gene and its coded protein neutral sphingomyelinase 2 (NSM) and alcohol abuse, emotional behavior, and bone defects were discovered and multiple mechanisms were identified for females. Here we report strong sex-dimorphisms for central, but not for peripheral mechanisms of NSM action in mouse models. Reduced NSM activity resulted in enhanced alcohol consumption in males, but delayed conditioned rewarding effects. It enhanced the acute dopamine response to alcohol, but decreased monoaminergic systems adaptations to chronic alcohol. Reduced NSM activity increased depression- and anxiety-like behavior, but was not involved in alcohol use for the self-management of the emotional state. Constitutively reduced NSM activity impaired structural development in the brain and enhanced lipidomic sensitivity to chronic alcohol. While the central effects were mostly opposite to NSM function in females, similar roles in bone-mediated osteocalcin release and its effects on alcohol drinking and emotional behavior were observed. These findings support the view that the NSM and multiple downstream mechanism may be a source of the sex-differences in alcohol use and emotional behavior.
Collapse
Affiliation(s)
- Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Tianye Jia
- The Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBI, Fudan University, Shanghai 200433, China.,PONS Centre and SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AB, UK
| | - Felix Anderheiden
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Maria Datz
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Anna-Lisa Eberle
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Volker Eulenburg
- Department for Anesthesiology and Intensive Care, Faculty of Medicine, University of Leipzig, Leipzig 04103, Germany
| | - Jonas Granzow
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Martin Hofer
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Julia Hohenschild
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Sabine E Huber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Stefanie Kämpf
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Georgios Kogias
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Laura Lacatusu
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Charlotte Lugmair
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Stephen Mbu Taku
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Doris Meixner
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Nina-Kristin Sembritzki
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Marc Praetner
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany.,Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich 82152, Germany
| | - Cosima Rhein
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Christina Sauer
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Jessica Scholz
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Franziska Ulrich
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Florian Valenta
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Esther Weigand
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Markus Werner
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Nicole Tay
- The Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBI, Fudan University, Shanghai 200433, China
| | - Conor J Mc Veigh
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Jana Haase
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - An-Li Wang
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf 40225, Germany
| | - Laila Abdel-Hafiz
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf 40225, Germany
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf 40225, Germany
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, Kraków 31-343, Poland
| | - Malgorzata Frankowska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, Kraków 31-343, Poland
| | - Malgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, Kraków 31-343, Poland
| | - Anbarasu Lourdusamy
- Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Philipp Kirchner
- Institute of Human Genetics, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen 91054, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen 91054, Germany
| | - Lena M Marx
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Neeraja Puliparambil Suresh
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Anna Fejtova
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Essa M Saied
- Institute for Chemistry, Humboldt University, Berlin 12489, Germany.,Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Christoph Arenz
- Institute for Chemistry, Humboldt University, Berlin 12489, Germany
| | - Aline Bozec
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen 91054, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen 91054, Germany
| | - Isabel Wank
- Department of Experimental and Clinical Pharmacology and Toxicology, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Silke Kreitz
- Department of Experimental and Clinical Pharmacology and Toxicology, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Andreas Hess
- Department of Experimental and Clinical Pharmacology and Toxicology, Emil Fischer Center, Friedrich-Alexander-University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Tobias Bäuerle
- Preclinical Imaging Platform Erlangen, Institute of Radiology, University Hospital Erlangen, Erlangen 91054, Germany
| | | | - Daniel N Mitroi
- Centro Biologia Molecular Severo Ochoa (CSIC-UAM), Madrid 28040, Spain
| | - André M Miranda
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Campus Gualtar, Braga 4710-057, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães 4710-057, Portugal
| | - Tiago Gil Oliveira
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Campus Gualtar, Braga 4710-057, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães 4710-057, Portugal
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany.,Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health (CIMH), Medical Faculty Mannheim, Heidelberg University, J5, Mannheim 68159, Germany
| | - Gunter Schumann
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany.,The Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBI, Fudan University, Shanghai 200433, China.,Department of Psychiatry and Psychotherapie, CCM, PONS Centre, Charite Mental Health, Charite Universitaetsmedizin Berlin, Berlin 10117, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, Erlangen 91054, Germany.,Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| |
Collapse
|
43
|
Surendran A, Ismail U, Atefi N, Bagchi AK, Singal PK, Shah A, Aliani M, Ravandi A. Lipidomic Predictors of Coronary No-Reflow. Metabolites 2023; 13:79. [PMID: 36677004 PMCID: PMC9861202 DOI: 10.3390/metabo13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
The ‘no-reflow’ phenomenon (NRP) after primary percutaneous coronary intervention (PCI) is a serious complication among acute ST-segment elevation myocardial infarction (STEMI) patients. Herein, a comprehensive lipidomics approach was used to quantify over 300 distinct molecular species in circulating plasma from 126 patients with STEMI before and after primary PCI. Our analysis showed that three lipid classes: phosphatidylcholine (PC), alkylphosphatidylcholine (PC(O)), and sphingomyelin (SM), were significantly elevated (p < 0.05) in no-reflow patients before primary PCI. The levels of individual fatty acids and total fatty acid levels were significantly lower (p < 0.05) in no-reflow subjects after PCI. The grouping of patients based on ECG ST-segment resolution (STR) also demonstrated the same trend, confirming the possible role of these differential lipids in the setting of no-reflow. Sphingomyelin species, SM 41:1 and SM 41:2, was invariably positively correlated with corrected TIMI frame count (CTFC) at pre-PCI and post-PCI. The plasma levels of SM 42:1 exhibited an inverse association (p < 0.05) consistently with tumor necrosis factor-alpha (TNF-α) at pre-PCI and post-PCI. In conclusion, we identified plasma lipid profiles that distinguish individuals at risk of no-reflow and provided novel insights into how dyslipidemia may contribute to NRP after primary PCI.
Collapse
Affiliation(s)
- Arun Surendran
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Umar Ismail
- Section of Cardiology, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Negar Atefi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Ashim K. Bagchi
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72204, USA
| | - Pawan K. Singal
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Ashish Shah
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Section of Cardiology, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michel Aliani
- Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Amir Ravandi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Section of Cardiology, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
44
|
Nutritional lipidomics for the characterization of lipids in food. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023. [PMID: 37516469 DOI: 10.1016/bs.afnr.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lipids represent one out of three major macronutrient classes in the human diet. It is estimated to account for about 15-20% of the total dietary intake. Triacylglycerides comprise the majority of them, estimated 90-95%. Other lipid classes include free fatty acids, phospholipids, cholesterol, and plant sterols as minor components. Various methods are used for the characterization of nutritional lipids, however, lipidomics approaches become increasingly attractive for this purpose due to their wide coverage, comprehensiveness and holistic view on composition. In this chapter, analytical methodologies and workflows utilized for lipidomics profiling of food samples are outlined with focus on mass spectrometry-based assays. The chapter describes common lipid extraction protocols, the distinct instrumental mass-spectrometry based analytical platforms for data acquisition, chromatographic and ion-mobility spectrometry methods for lipid separation, briefly mentions alternative methods such as gas chromatography for fatty acid profiling and mass spectrometry imaging. Critical issues of important steps of lipidomics workflows such as structural annotation and identification, quantification and quality assurance are discussed as well. Applications reported over the period of the last 5years are summarized covering the discovery of new lipids in foodstuff, differential profiling approaches for comparing samples from different origin, species, varieties, cultivars and breeds, and for food processing quality control. Lipidomics as a powerful tool for personalized nutrition and nutritional intervention studies is briefly discussed as well. It is expected that this field is significantly growing in the near future and this chapter gives a short insight into the power of nutritional lipidomics approaches.
Collapse
|
45
|
Borroni E, Frigerio G, Polledri E, Mercadante R, Maggioni C, Fedrizzi L, Pesatori AC, Fustinoni S, Carugno M. Metabolomic profiles in night shift workers: A cross-sectional study on hospital female nurses. Front Public Health 2023; 11:1082074. [PMID: 36908447 PMCID: PMC9999616 DOI: 10.3389/fpubh.2023.1082074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Background and aim Shift work, especially including night shifts, has been found associated with several diseases, including obesity, diabetes, cancers, and cardiovascular, mental, gastrointestinal and sleep disorders. Metabolomics (an omics-based methodology) may shed light on early biological alterations underlying these associations. We thus aimed to evaluate the effect of night shift work (NSW) on serum metabolites in a sample of hospital female nurses. Methods We recruited 46 nurses currently working in NSW in Milan (Italy), matched to 51 colleagues not employed in night shifts. Participants filled in a questionnaire on demographics, lifestyle habits, personal and family health history and work, and donated a blood sample. The metabolome was evaluated through a validated targeted approach measuring 188 metabolites. Only metabolites with at least 50% observations above the detection limit were considered, after standardization and log-transformation. Associations between each metabolite and NSW were assessed applying Tobit regression models and Random Forest, a machine-learning algorithm. Results When comparing current vs. never night shifters, we observed lower levels of 21 glycerophospholipids and 6 sphingolipids, and higher levels of serotonin (+171.0%, 95%CI: 49.1-392.7), aspartic acid (+155.8%, 95%CI: 40.8-364.7), and taurine (+182.1%, 95%CI: 67.6-374.9). The latter was higher in former vs. never night shifters too (+208.8%, 95%CI: 69.2-463.3). Tobit regression comparing ever (i.e., current + former) and never night shifters returned similar results. Years worked in night shifts did not seem to affect metabolite levels. The Random-Forest algorithm confirmed taurine and aspartic acid among the most important variables in discriminating current vs. never night shifters. Conclusions This study, although based on a small sample size, shows altered levels of some metabolites in night shift workers. If confirmed, our results may shed light on early biological alterations that might be related to adverse health effects of NSW.
Collapse
Affiliation(s)
- Elisa Borroni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Gianfranco Frigerio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Polledri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Rosa Mercadante
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Cristina Maggioni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Luca Fedrizzi
- Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Cecilia Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Fustinoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michele Carugno
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Occupational Health Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
46
|
Wang W, Song L. Landscape of lipidomics in cardiovascular medicine from 2012 to 2021: A systematic bibliometric analysis and literature review. Medicine (Baltimore) 2022; 101:e32599. [PMID: 36596038 PMCID: PMC9803420 DOI: 10.1097/md.0000000000032599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lipidomics has shaped our knowledge of how lipids play a central role in cardiovascular diseases (CVD), whereas there is a lack of a summary of existing research findings. This study performed a bibliometric analysis of lipidomics research in cardiovascular medicine to reveal the core countries, institutions, key researchers, important references, major journals, research hotspots and frontiers in this field. From 2012 to 2021, a total of 761 articles were obtained from the Web of Science Core Collection database. There is a steady increase of publications yearly. The United States and China are on the top of the list regarding article output. The institutions with the most publications were the Baker Heart and Diabetes Institute, the Chinese Academy of Sciences and Harvard Medical School. Peter J Meikle was both the most published and most co-cited author. The major journal in this field is Journal of lipid research. Keyword co-occurrence analysis indicated that coronary heart disease, mass spectrometry, risk, fatty acid, and insulin resistance have become hot topics in this field and keyword burst detection suggests that metabolomics, activation, liver, low density lipoprotein are the frontiers of research in recent years. Collectively, lipidomics in CVD is still in its infancy with a steady increase yearly. More in-depth studies in this area are warranted in the future.
Collapse
Affiliation(s)
- Wenting Wang
- Department of Cardiovascular Disease, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, Hangzhou, China
- * Correspondence: Wenting Wang, Department of Cardiology, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, 208 Huancheng East Road, Hangzhou 310003, China (e-mail: )
| | - Lei Song
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
47
|
Comparison of Workflows for Milk Lipid Analysis: Phospholipids. Foods 2022; 12:foods12010163. [PMID: 36613379 PMCID: PMC9818897 DOI: 10.3390/foods12010163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Milk is a rich source of lipids, with the major components being triglycerides (TAG) and phospholipids (mainly phosphatidylcholine (PC), sphingomyelin (SM), phosphatidylethanolamine (PE), phosphatidylserine (PS) and phosphatidylinositol (PI)). Liquid chromatography-mass spectrometry (LC-MS) is the predominant technique for lipid identification and quantification across all biological samples. While fatty acid (FA) composition of the major lipid classes of milk can be readily determined using tandem MS, elucidating the regio-distribution and double bond position of the FA remains difficult. Various workflows have been reported on the quantification of lipid species in biological samples in the past 20 years, but no standard or consensus methods are currently available for the quantification of milk phospholipids. This study will examine the influence of several common factors in lipid analysis workflow (including lipid extraction protocols, LC stationary phases, mobile phase buffers, gradient elution programmes, mass analyser resolution and isotope correction) on the quantification outcome of bovine milk phospholipids. The pros and cons of the current LC-MS methods as well as the critical problems to be solved will also be discussed.
Collapse
|
48
|
Li C, Qin D, Hu J, Yang Y, Hu D, Yu B. Inflamed adipose tissue: A culprit underlying obesity and heart failure with preserved ejection fraction. Front Immunol 2022; 13:947147. [PMID: 36483560 PMCID: PMC9723346 DOI: 10.3389/fimmu.2022.947147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
The incidence of heart failure with preserved ejection fraction is increasing in patients with obesity, diabetes, hypertension, and in the aging population. However, there is a lack of adequate clinical treatment. Patients with obesity-related heart failure with preserved ejection fraction display unique pathophysiological and phenotypic characteristics, suggesting that obesity could be one of its specific phenotypes. There has been an increasing recognition that overnutrition in obesity causes adipose tissue expansion and local and systemic inflammation, which consequently exacerbates cardiac remodeling and leads to the development of obese heart failure with preserved ejection fraction. Furthermore, overnutrition leads to cellular metabolic reprogramming and activates inflammatory signaling cascades in various cardiac cells, thereby promoting maladaptive cardiac remodeling. Growing evidence indicates that the innate immune response pathway from the NLRP3 inflammasome, to interleukin-1 to interleukin-6, is involved in the generation of obesity-related systemic inflammation and heart failure with preserved ejection fraction. This review established the existence of obese heart failure with preserved ejection fraction based on structural and functional changes, elaborated the inflammation mechanisms of obese heart failure with preserved ejection fraction, proposed that NLRP3 inflammasome activation may play an important role in adiposity-induced inflammation, and summarized the potential therapeutic approaches.
Collapse
Affiliation(s)
- Chenyu Li
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Jiarui Hu
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Die Hu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China
| | - Bilian Yu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, China,*Correspondence: Bilian Yu,
| |
Collapse
|
49
|
Saito K, Gemma A, Tatsumi K, Hattori N, Ushiki A, Tsushima K, Saito Y, Abe M, Horimasu Y, Kashiwada T, Mori K, Sato M, Nishiya T, Takamatsu K, Sun Y, Arakawa N, Izumi T, Ohno Y, Saito Y, Hanaoka M. Identification and characterization of lysophosphatidylcholine 14:0 as a biomarker for drug-induced lung disease. Sci Rep 2022; 12:19819. [PMID: 36396675 PMCID: PMC9671920 DOI: 10.1038/s41598-022-24406-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Drug-induced interstitial lung disease (DILD) occurs when drug exposure causes inflammation of the lung interstitium. DILD can be caused by different types of drugs, and some DILD patterns results in a high mortality rate; hence, DILD poses a serious problem in clinical practice as well as drug development, and strategies to diagnose and distinguish DILD from other lung diseases are necessary. We aimed to identify novel biomarkers for DILD by performing lipidomics analysis on plasma samples from patients with acute and recovery phase DILD. Having identified lysophosphatidylcholines (LPCs) as candidate biomarkers for DILD, we determined their concentrations using validated liquid chromatography/mass spectrometry biomarker assays. In addition, we evaluated the ability of LPCs to discriminate patients with acute phase DILD from those with recovery phase DILD, DILD-tolerant, or other lung diseases, and characterized their association with clinical characteristics. Lipidomics analysis revealed a clear decrease in LPC concentrations in the plasma of patients with acute phase DILD. In particular, LPC(14:0) had the highest discriminative index against recovery phase and DILD-tolerant patients. LPC(14:0) displayed no clear association with causal drugs, or subjects' backgrounds, but was associated with disease severity. Furthermore, LPC(14:0) was able to discriminate between patients with DILD and other lung diseases, including idiopathic interstitial pneumonia and lung disease associated with connective tissue disease. LPC(14:0) is a promising biomarker for DILD that could improve the diagnosis of DILD and help to differentiate DILD from other lung diseases, such as idiopathic interstitial pneumonia and connective tissue disease.
Collapse
Affiliation(s)
- Kosuke Saito
- grid.410797.c0000 0001 2227 8773Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, 210-9501 Japan
| | - Akihiko Gemma
- grid.410821.e0000 0001 2173 8328Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603 Japan
| | - Koichiro Tatsumi
- grid.136304.30000 0004 0370 1101Department of Respirology (B2), Graduate School of Medicine, Chiba University, Chiba, 260-8677 Japan
| | - Noboru Hattori
- grid.470097.d0000 0004 0618 7953Department of Respiratory Medicine, Hiroshima University Hospital, Hiroshima, 734-8551 Japan
| | - Atsuhito Ushiki
- grid.263518.b0000 0001 1507 4692First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, 390-8621 Japan
| | - Kenji Tsushima
- grid.410797.c0000 0001 2227 8773Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, 210-9501 Japan ,grid.411731.10000 0004 0531 3030School of Medicine, International University of Health and Welfare, Narita, 286-8686 Japan
| | - Yoshinobu Saito
- grid.410821.e0000 0001 2173 8328Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603 Japan
| | - Mitsuhiro Abe
- grid.136304.30000 0004 0370 1101Department of Respirology (B2), Graduate School of Medicine, Chiba University, Chiba, 260-8677 Japan
| | - Yasushi Horimasu
- grid.470097.d0000 0004 0618 7953Department of Respiratory Medicine, Hiroshima University Hospital, Hiroshima, 734-8551 Japan
| | - Takeru Kashiwada
- grid.410821.e0000 0001 2173 8328Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603 Japan
| | - Kazuhiko Mori
- grid.410844.d0000 0004 4911 4738Daiichi Sankyo RD Novare Co., Ltd., Tokyo, 134-8630 Japan
| | - Motonobu Sato
- grid.418042.b0000 0004 1758 8699Astellas Pharma Inc., Tsukuba, 305-8585 Japan
| | - Takayoshi Nishiya
- grid.410844.d0000 0004 4911 4738Daiichi Sankyo RD Novare Co., Ltd., Tokyo, 134-8630 Japan
| | - Kazuhiko Takamatsu
- grid.418042.b0000 0004 1758 8699Astellas Pharma Inc., Tsukuba, 305-8585 Japan
| | - Yuchen Sun
- grid.410797.c0000 0001 2227 8773Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, 210-9501 Japan
| | - Noriaki Arakawa
- grid.410797.c0000 0001 2227 8773Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, 210-9501 Japan
| | - Takashi Izumi
- Kihara Memorial Foundation, Yokohama, 230-0045 Japan
| | - Yasuo Ohno
- Kihara Memorial Foundation, Yokohama, 230-0045 Japan
| | - Yoshiro Saito
- grid.410797.c0000 0001 2227 8773Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, 210-9501 Japan
| | - Masayuki Hanaoka
- grid.263518.b0000 0001 1507 4692First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, 390-8621 Japan
| |
Collapse
|
50
|
Zeng W, Beyene HB, Kuokkanen M, Miao G, Magliano DJ, Umans JG, Franceschini N, Cole SA, Michailidis G, Lee ET, Howard BV, Fiehn O, Curran JE, Blangero J, Meikle PJ, Zhao J. Lipidomic profiling in the Strong Heart Study identified American Indians at risk of chronic kidney disease. Kidney Int 2022; 102:1154-1166. [PMID: 35853479 PMCID: PMC10753995 DOI: 10.1016/j.kint.2022.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022]
Abstract
Dyslipidemia associates with and usually precedes the onset of chronic kidney disease (CKD), but a comprehensive assessment of molecular lipid species associated with risk of CKD is lacking. Here, we sought to identify fasting plasma lipids associated with risk of CKD among American Indians in the Strong Heart Family Study, a large-scale community-dwelling of individuals, followed by replication in Mexican Americans from the San Antonio Family Heart Study and Caucasians from the Australian Diabetes, Obesity and Lifestyle Study. We also performed repeated measurement analysis to examine the temporal relationship between the change in the lipidome and change in kidney function between baseline and follow-up of about five years apart. Network analysis was conducted to identify differential lipid classes associated with risk of CKD. In the discovery cohort, we found that higher baseline level of multiple lipid species, including glycerophospholipids, glycerolipids and sphingolipids, was significantly associated with increased risk of CKD, independent of age, sex, body mass index, diabetes and hypertension. Many lipid species were replicated in at least one external cohort at the individual lipid species and/or the class level. Longitudinal change in the plasma lipidome was significantly associated with change in the estimated glomerular filtration rate after adjusting for covariates, baseline lipids and the baseline rate. Network analysis identified distinct lipidomic signatures differentiating high from low-risk groups. Thus, our results demonstrated that disturbed lipid metabolism precedes the onset of CKD. These findings shed light on the mechanisms linking dyslipidemia to CKD and provide potential novel biomarkers for identifying individuals with early impaired kidney function at preclinical stages.
Collapse
Affiliation(s)
- Wenjie Zeng
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mikko Kuokkanen
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | - Guanhong Miao
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Jason G Umans
- MedStar Health Research Institute, Hyattsville, Maryland, USA; Georgetown-Howard Universities Center for Clinical and Translational Science, Washington, District of Columbia, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - George Michailidis
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Elisa T Lee
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, Maryland, USA; Georgetown-Howard Universities Center for Clinical and Translational Science, Washington, District of Columbia, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California-Davis, Davis, California, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|