1
|
Skotland T, Ekroos K, Llorente A, Sandvig K. Quantitative Lipid Analysis of Extracellular Vesicle Preparations: A Perspective. J Extracell Vesicles 2025; 14:e70049. [PMID: 40091364 PMCID: PMC11911390 DOI: 10.1002/jev2.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Quantitative lipidomic analysis performed by mass spectrometry is required for determination of the lipid content of extracellular vesicles (EVs). Such methods can provide information about the total amount of lipids, the lipid species composition, the purity of EV samples as well as the cellular origin of the EVs. There are, however, many pitfalls when performing lipid analyses. Thus, any non-specialist should collaborate with experts in lipidomics. In addition to many good review articles giving advice about lipid analyses, we recommend the information and guidelines published by the Lipidomic Standard Initiative, an interest group affiliated with the International Lipidomics Society.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Centre for Cancer Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kim Ekroos
- Lipidomic Consulting Ltd., Espoo, Finland
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Centre for Cancer Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Centre for Cancer Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of diet and genotype on the lipidome of mice with altered lipoprotein metabolism. iScience 2024; 27:111051. [PMID: 39568621 PMCID: PMC11577568 DOI: 10.1016/j.isci.2024.111051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
The present study describes and compares the impact of PCSK9 and LDLR, two pivotal players in cholesterol metabolism, on the whole lipidome of plasma, liver and aorta in different dietary conditions. This issue is relevant, since several lipid species, circulating at very low concentrations, have the ability to impair lipid metabolism and promote atherosclerosis development. To this aim, wild-type, hypercholesterolemic Ldlr-KO, and hypocholesterolemic Pcsk9-KO mice were fed a standard chow or a Western-type diet up to 30 and 16 weeks of age, respectively. 42 lipids including cholesterol, cholesteryl esters, several sphingolipids, phospholipids, and lysophospholipids, accumulated uniquely in the atherosclerotic aorta of Western-type diet-fed Ldlr-KO mice. In addition, multiple organ/tissue comparisons allowed us to identify 16 lipids whose plasma and hepatic patterns mirrored the lipidome of the atherosclerotic aorta. These lipid species, belonging to cholesteryl esters, glucosyl/galactosylceramide, lactosylceramide, globotriaosylceramide, sphingomyelin, and phosphatidylcholine could be further investigated as circulating biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, via Balzaretti, 9, Milan, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, via Balzaretti, 9, Milan, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, via Balzaretti, 9, Milan, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, via Balzaretti, 9, Milan, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, 02150 Espoo, Finland
- Finnish Cardiovascular Research Center, University of Tampere, 33520 Tampere, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, via Balzaretti, 9, Milan, Italy
| |
Collapse
|
3
|
Martínez-Díaz P, Parra A, Sanchez-López CM, Casas J, Lucas X, Marcilla A, Roca J, Barranco I. Small and Large Extracellular Vesicles of Porcine Seminal Plasma Differ in Lipid Profile. Int J Mol Sci 2024; 25:7492. [PMID: 39000599 PMCID: PMC11242203 DOI: 10.3390/ijms25137492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Seminal plasma contains a heterogeneous population of extracellular vesicles (sEVs) that remains poorly characterized. This study aimed to characterize the lipidomic profile of two subsets of differently sized sEVs, small (S-) and large (L-), isolated from porcine seminal plasma by size-exclusion chromatography and characterized by an orthogonal approach. High-performance liquid chromatography-high-resolution mass spectrometry was used for lipidomic analysis. A total of 157 lipid species from 14 lipid classes of 4 major categories (sphingolipids, glycerophospholipids, glycerolipids, and sterols) were identified. Qualitative differences were limited to two cholesteryl ester species present only in S-sEVs. L-sEVs had higher levels of all quantified lipid classes due to their larger membrane surface area. The distribution pattern was different, especially for sphingomyelins (more in S-sEVs) and ceramides (more in L-sEVs). In conclusion, this study reveals differences in the lipidomic profile of two subsets of porcine sEVs, suggesting that they differ in biogenesis and functionality.
Collapse
Affiliation(s)
- Pablo Martínez-Díaz
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Ana Parra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Christian M Sanchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46100 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de València, 46100 Valencia, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Institute for Advanced Chemistry (IQAC-CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Xiomara Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, 46100 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de València, 46100 Valencia, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| | - Isabel Barranco
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, 30100 Murcia, Spain
| |
Collapse
|
4
|
Sharma S, Subrahmanyam YV, Ranjani H, Sidra S, Parmar D, Vadivel S, Kannan S, Grallert H, Usharani D, Anjana RM, Balasubramanyam M, Mohan V, Jerzy A, Panchagnula V, Gokulakrishnan K. Circulatory levels of lysophosphatidylcholine species in obese adolescents: Findings from cross-sectional and prospective lipidomics analyses. Nutr Metab Cardiovasc Dis 2024; 34:1807-1816. [PMID: 38503619 DOI: 10.1016/j.numecd.2024.02.009] [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: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND AND AIMS Obesity has reached epidemic proportions, emphasizing the importance of reliable biomarkers for detecting early metabolic alterations and enabling early preventative interventions. However, our understanding of the molecular mechanisms and specific lipid species associated with childhood obesity remains limited. Therefore, the aim of this study was to investigate plasma lipidomic signatures as potential biomarkers for adolescent obesity. METHODS AND RESULTS A total of 103 individuals comprising overweight/obese (n = 46) and normal weight (n = 57) were randomly chosen from the baseline ORANGE (Obesity Reduction and Noncommunicable Disease Awareness through Group Education) cohort, having been followed up for a median of 7.1 years. Plasma lipidomic profiling was performed using the UHPLC-HRMS method. We used three different models adjusted for clinical covariates to analyze the data. Clustering methods were used to define metabotypes, which allowed for the stratification of subjects into subgroups with similar clinical and metabolic profiles. We observed that lysophosphatidylcholine (LPC) species like LPC.16.0, LPC.18.3, LPC.18.1, and LPC.20.3 were significantly (p < 0.05) associated with baseline and follow-up BMI in adolescent obesity. The association of LPC species with BMI remained consistently significant even after adjusting for potential confounders. Moreover, applying metabotyping using hierarchical clustering provided insights into the metabolic heterogeneity within the normal and obese groups, distinguishing metabolically healthy individuals from those with unhealthy metabolic profiles. CONCLUSION The specific LPC levels were found to be altered and increased in childhood obesity, particularly during the follow-up. These findings suggest that LPC species hold promise as potential biomarkers of obesity in adolescents, including healthy and unhealthy metabolic profiles.
Collapse
Affiliation(s)
- Sapna Sharma
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Yalamanchili Venkata Subrahmanyam
- CEPD Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008 India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harish Ranjani
- Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India; Department of Preventive and Digital Health Research, Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India
| | - Sidra Sidra
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Dharmeshkumar Parmar
- CEPD Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008 India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sangeetha Vadivel
- Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India
| | - Shanthini Kannan
- Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Dandamudi Usharani
- Department of Food Safety and Analytical Quality Control Laboratory, CSIR-Central Food Technological Research Institute (CFTRI), Mysore, Karnataka 570020, India
| | - Ranjit Mohan Anjana
- Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India
| | | | - Viswanathan Mohan
- Madras Diabetes Research Foundation, No. 4, Conran Smith Road, Gopalapuram, Chennai, 600086 India
| | - Adamski Jerzy
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, 117597, Singapore; Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Venkateswarlu Panchagnula
- CEPD Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008 India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuppan Gokulakrishnan
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, Karnataka 560029, India.
| |
Collapse
|
5
|
Gu TJ, Liu PK, Wang YW, Flowers MT, Xu S, Liu Y, Davis DB, Li L. Diazobutanone-assisted isobaric labelling of phospholipids and sulfated glycolipids enables multiplexed quantitative lipidomics using tandem mass spectrometry. Nat Chem 2024; 16:762-770. [PMID: 38365942 DOI: 10.1038/s41557-023-01436-2] [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: 01/10/2023] [Accepted: 12/21/2023] [Indexed: 02/18/2024]
Abstract
Mass spectrometry-based quantitative lipidomics is an emerging field aiming to uncover the intricate relationships between lipidomes and disease development. However, quantifying lipidomes comprehensively in a high-throughput manner remains challenging owing to the diverse lipid structures. Here we propose a diazobutanone-assisted isobaric labelling strategy as a rapid and robust platform for multiplexed quantitative lipidomics across a broad range of lipid classes, including various phospholipids and glycolipids. The diazobutanone reagent is designed to conjugate with phosphodiester or sulfate groups, while accommodating various functional groups on different lipid classes, enabling subsequent isobaric labelling for high-throughput multiplexed quantitation. Our method demonstrates excellent performance in terms of labelling efficiency, detection sensitivity, quantitative accuracy and broad applicability to various biological samples. Finally, we performed a six-plex quantification analysis of lipid extracts from lean and obese mouse livers. In total, we identified and quantified 246 phospholipids in a high-throughput manner, revealing lipidomic changes that may be associated with obesity in mice.
Collapse
Affiliation(s)
- Ting-Jia Gu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Peng-Kai Liu
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yen-Wen Wang
- Department of Biostatics, Yale University, New Haven, CT, USA
| | - Matthew T Flowers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Shuling Xu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuan Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Dawn B Davis
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
6
|
Zhang J, Ruiz M, Bergh PO, Henricsson M, Stojanović N, Devkota R, Henn M, Bohlooly-Y M, Hernández-Hernández A, Alsheimer M, Borén J, Pilon M, Shibuya H. Regulation of meiotic telomere dynamics through membrane fluidity promoted by AdipoR2-ELOVL2. Nat Commun 2024; 15:2315. [PMID: 38485951 PMCID: PMC10940294 DOI: 10.1038/s41467-024-46718-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
The cellular membrane in male meiotic germ cells contains a unique class of phospholipids and sphingolipids that is required for male reproduction. Here, we show that a conserved membrane fluidity sensor, AdipoR2, regulates the meiosis-specific lipidome in mouse testes by promoting the synthesis of sphingolipids containing very-long-chain polyunsaturated fatty acids (VLC-PUFAs). AdipoR2 upregulates the expression of a fatty acid elongase, ELOVL2, both transcriptionally and post-transcriptionally, to synthesize VLC-PUFA. The depletion of VLC-PUFAs and subsequent accumulation of palmitic acid in AdipoR2 knockout testes stiffens the cellular membrane and causes the invagination of the nuclear envelope. This condition impairs the nuclear peripheral distribution of meiotic telomeres, leading to errors in homologous synapsis and recombination. Further, the stiffened membrane impairs the formation of intercellular bridges and the germ cell syncytium, which disrupts the orderly arrangement of cell types within the seminiferous tubules. According to our findings we propose a framework in which the highly-fluid membrane microenvironment shaped by AdipoR2-ELOVL2 underpins meiosis-specific chromosome dynamics in testes.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Per-Olof Bergh
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, 41467, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, 41467, Gothenburg, Sweden
| | - Nena Stojanović
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Ranjan Devkota
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Marius Henn
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | | | - Abrahan Hernández-Hernández
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- National Genomics Infrastructure, Science for Life Laboratory, Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Manfred Alsheimer
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, 41467, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden.
| | - Hiroki Shibuya
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden.
- Laboratory for Gametogenesis, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan.
| |
Collapse
|
7
|
Merciai F, Basilicata MG, La Gioia D, Salviati E, Caponigro V, Ciaglia T, Musella S, Crescenzi C, Sommella E, Campiglia P. Sub-5-min RP-UHPLC-TIMS for high-throughput untargeted lipidomics and its application to multiple matrices. Anal Bioanal Chem 2024; 416:959-970. [PMID: 38078946 DOI: 10.1007/s00216-023-05084-w] [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: 09/20/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 01/23/2024]
Abstract
Untargeted lipidomics, with its ability to take a snapshot of the lipidome landscape, is an important tool to highlight lipid changes in pathology or drug treatment models. One of the shortcomings of most untargeted lipidomics based on UHPLC-HRMS is the low throughput, which is not compatible with large-scale screening. In this contribution, we evaluate the application of a sub-5-min high-throughput four-dimensional trapped ion mobility mass spectrometry (HT-4D-TIMS) platform for the fast profiling of multiple complex biological matrices. Human AC-16 cells and mouse brain, liver, sclera, and feces were used as samples. By using a fast 4-min RP gradient, the implementation of TIMS allows us to differentiate coeluting isomeric and isobaric lipids, with correct precursor ion isolation, avoiding co-fragmentation and chimeric MS/MS spectra. Globally, the HT-4D-TIMS allowed us to annotate 1910 different lipid species, 1308 at the molecular level and 602 at the sum composition level, covering 58 lipid subclasses, together with quantitation capability covering more than three orders of magnitude. Notably, TIMS values were highly comparable with respect to longer LC gradients (CV% = 0.39%). These results highlight how HT-4D-TIMS-based untargeted lipidomics possess high coverage and accuracy, halving the analysis time with respect to conventional UHPLC methods, and can be used for fast and accurate untargeted analysis of complex matrices to rapidly evaluate changes of lipid metabolism in disease models or drug discovery campaigns.
Collapse
Affiliation(s)
- Fabrizio Merciai
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | | | - Danila La Gioia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
- PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | - Vicky Caponigro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | - Carlo Crescenzi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy.
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 13284084, Fisciano, SA, Italy
| |
Collapse
|
8
|
Skotland T, Llorente A, Sandvig K. Lipids in Extracellular Vesicles: What Can Be Learned about Membrane Structure and Function? Cold Spring Harb Perspect Biol 2023; 15:a041415. [PMID: 37277192 PMCID: PMC10411865 DOI: 10.1101/cshperspect.a041415] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular vesicles, such as exosomes, can be used as interesting models to study the structure and function of biological membranes as these vesicles contain only one membrane (i.e., one lipid bilayer). In addition to lipids, they contain proteins, nucleic acids, and various other molecules. The lipid composition of exosomes is here compared to HIV particles and detergent-resistant membranes, which also have a high content of sphingolipids, cholesterol, and phosphatidylserine (PS). We discuss interactions between the lipids in the two bilayers, and especially those between PS 18:0/18:1 in the inner leaflet and the very-long-chain sphingolipids in the outer leaflet, and the importance of cholesterol for these interactions. We also briefly discuss the involvement of ether-linked phospholipids (PLs) in such lipid raft-like structures, and the possible involvement of these and other lipid classes in the formation of exosomes. The urgent need to improve the quality of quantitative lipidomic studies is highlighted.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
9
|
Saha S, Singh P, Dutta A, Vaidya H, Negi PC, Sengupta S, Seth S, Basak T. A Comprehensive Insight and Mechanistic Understanding of the Lipidomic Alterations Associated With DCM. JACC. ASIA 2023; 3:539-555. [PMID: 37614533 PMCID: PMC10442885 DOI: 10.1016/j.jacasi.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 08/25/2023]
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure characterized by the enlargement of the left ventricular cavity and contractile dysfunction of the myocardium. Lipids are the major sources of energy for the myocardium. Impairment of lipid homeostasis has a potential role in the pathogenesis of DCM. In this review, we have summarized the role of different lipids in the progression of DCM that can be considered as potential biomarkers. Further, we have also explained the mechanistic pathways followed by the lipid molecules in disease progression along with the cardioprotective role of certain lipids. As the global epidemiological status of DCM is alarming, it is high time to define some disease-specific biomarkers with greater prognostic value. We are proposing an adaptation of a system lipidomics-based approach to profile DCM patients in order to achieve a better diagnosis and prognosis of the disease.
Collapse
Affiliation(s)
- Shubham Saha
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Abhi Dutta
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Hiteshi Vaidya
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Prakash Chand Negi
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| |
Collapse
|
10
|
Ruiz M, Devkota R, Kaper D, Ruhanen H, Busayavalasa K, Radović U, Henricsson M, Käkelä R, Borén J, Pilon M. AdipoR2 recruits protein interactors to promote fatty acid elongation and membrane fluidity. J Biol Chem 2023:104799. [PMID: 37164154 DOI: 10.1016/j.jbc.2023.104799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/12/2023] Open
Abstract
The human AdipoR2 and its C. elegans homolog PAQR-2 are multi-pass plasma membrane proteins that protect cells against membrane rigidification. However, how AdipoR2 promotes membrane fluidity mechanistically is not clear. Using 13C-labelled fatty acids, we show that AdipoR2 can promote the elongation and incorporation of membrane-fluidizing polyunsaturated fatty acids into phospholipids. To elucidate the molecular basis of these activities, we performed immunoprecipitations of tagged AdipoR2 and PAQR-2 expressed in HEK293 cells or whole C. elegans, respectively, and identified co-immunoprecipitated proteins using mass spectroscopy. We found that several of the evolutionarily conserved AdipoR2/PAQR-2 interactors are important for fatty acid elongation and incorporation into phospholipids. We experimentally verified some of these interactions, namely with the dehydratase HACD3 that is essential for the third of four steps in long-chain fatty acid elongation, and ACSL4 that is important for activation of unsaturated fatty acids and their channeling into phospholipids. We conclude that AdipoR2 and PAQR-2 can recruit protein interactors to promote the production and incorporation of unsaturated fatty acids into phospholipids.
Collapse
Affiliation(s)
- Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Ranjan Devkota
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Delaney Kaper
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science, Biocenter Finland, Helsinki, Finland; Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Kiran Busayavalasa
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Uroš Radović
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science, Biocenter Finland, Helsinki, Finland; Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
11
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of Diets on Plasma and Aorta Lipidome: A Study in the apoE Knockout Mouse Model. Mol Nutr Food Res 2023; 67:e2200367. [PMID: 36419336 DOI: 10.1002/mnfr.202200367] [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: 06/06/2022] [Revised: 09/20/2022] [Indexed: 11/27/2022]
Abstract
SCOPE Specific lipid molecules circulating in plasma at low concentrations have emerged as biomarkers of atherosclerotic risk. The aim of the present study is that of evaluating, in an athero-prone mouse model, how different diets can affect plasma and aorta lipidome. METHODS AND RESULTS Thirty-six apoE knockout mice are divided in three groups and feed 12 weeks with diets differing for cholesterol and fatty acid content. Atherosclerosis is measured at the aortic sinus and aorta. Lipids are quantified in plasma and aorta with mass spectrometry. The cholesterol content of the diets is the main driver of lipid accumulation in plasma and aorta. The fatty acid composition of the diets affects plasma levels both of essential (linoleic acid) and nonessential (myristic and arachidonic acid) ones. Lipidomics show a comparable distribution, in plasma and aorta, of the main lipid components of oxidized LDL, including cholesteryl esters and lysophosphatidylcholines. Interestingly, lactosylceramide, glucosyl/galactosylceramide, and individual ceramide species are found to accumulate in diseased aortic segments. CONCLUSION Both the cholesterol and fatty acid content of the diets profoundly affect plasma lipidome. Aorta lipidome is likewise affected with the accumulation of specific lipids known as markers of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, 02150, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, 33520, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| |
Collapse
|
12
|
Sphingosine 1-phosphate mediates adiponectin receptor signaling essential for lipid homeostasis and embryogenesis. Nat Commun 2022; 13:7162. [PMID: 36418331 PMCID: PMC9684441 DOI: 10.1038/s41467-022-34931-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Cells and organisms require proper membrane composition to function and develop. Phospholipids are the major component of membranes and are primarily acquired through the diet. Given great variability in diet composition, cells must be able to deploy mechanisms that correct deviations from optimal membrane composition and properties. Here, using lipidomics and unbiased proteomics, we found that the embryonic lethality in mice lacking the fluidity regulators Adiponectin Receptors 1 and 2 (AdipoR1/2) is associated with aberrant high saturation of the membrane phospholipids. Using mouse embryonic fibroblasts (MEFs) derived from AdipoR1/2-KO embryos, human cell lines and the model organism C. elegans we found that, mechanistically, AdipoR1/2-derived sphingosine 1-phosphate (S1P) signals in parallel through S1PR3-SREBP1 and PPARγ to sustain the expression of the fatty acid desaturase SCD and maintain membrane properties. Thus, our work identifies an evolutionary conserved pathway by which cells and organisms achieve membrane homeostasis and adapt to a variable environment.
Collapse
|
13
|
Need for more focus on lipid species in studies of biological and model membranes. Prog Lipid Res 2022; 86:101160. [DOI: 10.1016/j.plipres.2022.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/06/2022] [Indexed: 11/23/2022]
|
14
|
Vaura F, Palmu J, Aittokallio J, Kauko A, Niiranen T. Genetic, Molecular, and Cellular Determinants of Sex-Specific Cardiovascular Traits. Circ Res 2022; 130:611-631. [PMID: 35175841 DOI: 10.1161/circresaha.121.319891] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the well-known sex dimorphism in cardiovascular disease traits, the exact genetic, molecular, and cellular underpinnings of these differences are not well understood. A growing body of evidence currently points at the links between cardiovascular disease traits and the genome, epigenome, transcriptome, and metabolome. However, the sex-specific differences in these links remain largely unstudied due to challenges in bioinformatic methods, inadequate statistical power, analytic costs, and paucity of valid experimental models. This review article provides an overview of the literature on sex differences in genetic architecture, heritability, epigenetic changes, transcriptomic signatures, and metabolomic profiles in relation to cardiovascular disease traits. We also review the literature on the associations between sex hormones and cardiovascular disease traits and discuss the potential mechanisms underlying these associations, focusing on human studies.
Collapse
Affiliation(s)
- Felix Vaura
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Joonatan Palmu
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Jenni Aittokallio
- Department of Anesthesiology and Intensive Care (J.A.), University of Turku, Finland.,Division of Perioperative Services, Intensive Care and Pain Medicine (J.A.), Turku University Hospital, Finland
| | - Anni Kauko
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland.,Division of Medicine (T.N.), Turku University Hospital, Finland.,Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland (T.N.)
| |
Collapse
|
15
|
Ruiz M, Henricsson M, Borén J, Pilon M. Palmitic acid causes increased dihydroceramide levels when desaturase expression is directly silenced or indirectly lowered by silencing AdipoR2. Lipids Health Dis 2021; 20:173. [PMID: 34839823 PMCID: PMC8627610 DOI: 10.1186/s12944-021-01600-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background AdipoR1 and AdipoR2 (AdipoRs) are plasma membrane proteins often considered to act as adiponectin receptors with a ceramidase activity. Additionally, the AdipoRs and their yeast and C. elegans orthologs are emerging as membrane homeostasis regulators that counter membrane rigidification by promoting fatty acid desaturation and incorporation of unsaturated fatty acids into phospholipids, thus restoring fluidity. Methods Using cultured cells, the effects of AdipoR silencing or over-expression on the levels and composition of several sphingolipid classes were examined. Results AdipoR2 silencing in the presence of exogenous palmitic acid potently causes increased levels of dihydroceramides, a ceramide precursor in the de novo ceramide synthesis pathway. Conversely, AdipoR2 over-expression caused a depletion of dihydroceramides. Conclusions The results are consistent with AdipoR2 silencing leading to increased intracellular supply of palmitic acid that in turn leads to increased dihydroceramide synthesis via the rate-limiting serine palmitoyl transferase step. In agreement with this model, inhibiting the desaturase SCD or SREBF1/2 (positive regulators of SCD) also causes a strong increase in dihydroceramide levels. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01600-y.
Collapse
Affiliation(s)
- Mario Ruiz
- Department Chemistry and Molecular Biology, Univ. Gothenburg, Box 462, 405 30, Gothenburg, Sweden
| | - Marcus Henricsson
- Department Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30, Gothenburg, Sweden
| | - Jan Borén
- Department Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30, Gothenburg, Sweden
| | - Marc Pilon
- Department Chemistry and Molecular Biology, Univ. Gothenburg, Box 462, 405 30, Gothenburg, Sweden.
| |
Collapse
|
16
|
Meikle TG, Huynh K, Giles C, Meikle PJ. Clinical lipidomics: realizing the potential of lipid profiling. J Lipid Res 2021; 62:100127. [PMID: 34582882 PMCID: PMC8528718 DOI: 10.1016/j.jlr.2021.100127] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022] Open
Abstract
Dysregulation of lipid metabolism plays a major role in the etiology and sequelae of inflammatory disorders, cardiometabolic and neurological diseases, and several forms of cancer. Recent advances in lipidomic methodology allow comprehensive lipidomic profiling of clinically relevant biological samples, enabling researchers to associate lipid species and metabolic pathways with disease onset and progression. The resulting data serve not only to advance our fundamental knowledge of the underlying disease process but also to develop risk assessment models to assist in the diagnosis and management of disease. Currently, clinical applications of in-depth lipidomic profiling are largely limited to the use of research-based protocols in the analysis of population or clinical sample sets. However, we foresee the development of purpose-built clinical platforms designed for continuous operation and clinical integration-assisting health care providers with disease risk assessment, diagnosis, and monitoring. Herein, we review the current state of clinical lipidomics, including the use of research-based techniques and platforms in the analysis of clinical samples as well as assays already available to clinicians. With a primary focus on MS-based strategies, we examine instrumentation, analysis techniques, statistical models, prospective design of clinical platforms, and the possible pathways toward implementation of clinical lipidomics.
Collapse
Affiliation(s)
- Thomas G Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Corey Giles
- Metabolomics 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; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
17
|
Devkota R, Kaper D, Bodhicharla R, Henricsson M, Borén J, Pilon M. A genetic titration of membrane composition in Caenorhabditis elegans reveals its importance for multiple cellular and physiological traits. Genetics 2021; 219:iyab093. [PMID: 34125894 PMCID: PMC9335940 DOI: 10.1093/genetics/iyab093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022] Open
Abstract
Communicating editor: B. Grant The composition and biophysical properties of cellular membranes must be tightly regulated to maintain the proper functions of myriad processes within cells. To better understand the importance of membrane homeostasis, we assembled a panel of five Caenorhabditis elegans strains that show a wide span of membrane composition and properties, ranging from excessively rich in saturated fatty acids (SFAs) and rigid to excessively rich in polyunsaturated fatty acids (PUFAs) and fluid. The genotypes of the five strain are, from most rigid to most fluid: paqr-1(tm3262); paqr-2(tm3410), paqr-2(tm3410), N2 (wild-type), mdt-15(et14); nhr-49(et8), and mdt-15(et14); nhr-49(et8); acs-13(et54). We confirmed the excess SFA/rigidity-to-excess PUFA/fluidity gradient using the methods of fluorescence recovery after photobleaching (FRAP) and lipidomics analysis. The five strains were then studied for a variety of cellular and physiological traits and found to exhibit defects in: permeability, lipid peroxidation, growth at different temperatures, tolerance to SFA-rich diets, lifespan, brood size, vitellogenin trafficking, oogenesis, and autophagy during starvation. The excessively rigid strains often exhibited defects in opposite directions compared to the excessively fluid strains. We conclude that deviation from wild-type membrane homeostasis is pleiotropically deleterious for numerous cellular/physiological traits. The strains introduced here should prove useful to further study the cellular and physiological consequences of impaired membrane homeostasis.
Collapse
Affiliation(s)
- Ranjan Devkota
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg S-405 30, Sweden
| | - Delaney Kaper
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg S-405 30, Sweden
| | - Rakesh Bodhicharla
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg S-405 30, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg S-405 30, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg S-405 30, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg S-405 30, Sweden
| |
Collapse
|
18
|
Sas KM, Lin J, Wang CH, Zhang H, Saha J, Rajendiran TM, Soni T, Nair V, Eichinger F, Kretzler M, Brosius FC, Michailidis G, Pennathur S. Renin-angiotensin system inhibition reverses the altered triacylglycerol metabolic network in diabetic kidney disease. Metabolomics 2021; 17:65. [PMID: 34219205 PMCID: PMC8312633 DOI: 10.1007/s11306-021-01816-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Dyslipidemia is a significant risk factor for progression of diabetic kidney disease (DKD). Determining the changes in individual lipids and lipid networks across a spectrum of DKD severity may identify lipids that are pathogenic to DKD progression. METHODS We performed untargeted lipidomic analysis of kidney cortex tissue from diabetic db/db and db/db eNOS-/- mice along with non-diabetic littermate controls. A subset of mice were treated with the renin-angiotensin system (RAS) inhibitors, lisinopril and losartan, which improves the DKD phenotype in the db/db eNOS-/- mouse model. RESULTS Of the three independent variables in this study, diabetes had the largest impact on overall lipid levels in the kidney cortex, while eNOS expression and RAS inhibition had smaller impacts on kidney lipid levels. Kidney lipid network architecture, particularly of networks involving glycerolipids such as triacylglycerols, was substantially disrupted by worsening kidney disease in the db/db eNOS-/- mice compared to the db/db mice, a feature that was reversed with RAS inhibition. This was associated with decreased expression of the stearoyl-CoA desaturases, Scd1 and Scd2, with RAS inhibition. CONCLUSIONS In addition to the known salutary effect of RAS inhibition on DKD progression, our results suggest a previously unrecognized role for RAS inhibition on the kidney triacylglycerol lipid metabolic network.
Collapse
Affiliation(s)
- Kelli M Sas
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Jiahe Lin
- Department of Statistics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Chih-Hong Wang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Hongyu Zhang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Jharna Saha
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Thekkelnaycke M Rajendiran
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Felix Eichinger
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Frank C Brosius
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Division of Nephrology, Department of Medicine, University of Arizona, Tucson, Arizona, 85724, USA
| | - George Michailidis
- Department of Statistics and Computer and Information Sciences, University of Florida, Gainesville, Florida, 32611, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA.
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
19
|
The Protein Toxins Ricin and Shiga Toxin as Tools to Explore Cellular Mechanisms of Internalization and Intracellular Transport. Toxins (Basel) 2021; 13:toxins13060377. [PMID: 34070659 PMCID: PMC8227415 DOI: 10.3390/toxins13060377] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/22/2021] [Indexed: 12/18/2022] Open
Abstract
Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.
Collapse
|
20
|
Yi X, Li Y, Hu X, Wang F, Liu T. Changes in phospholipid metabolism in exosomes of hormone-sensitive and hormone-resistant prostate cancer cells. J Cancer 2021; 12:2893-2902. [PMID: 33854590 PMCID: PMC8040901 DOI: 10.7150/jca.48906] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 03/04/2021] [Indexed: 01/05/2023] Open
Abstract
Background: To explore the changes in lipids in exosomes of hormone-sensitive and hormone-resistant prostate cancer cells and develop an inexpensive and rapid technique for screening lipid-based biomarkers of prostate cancer. Methods: Exosomes were extracted from LnCap, PC3 and DU-145 cells, and their lipid composition was analyzed quantitatively using high-throughput mass spectrometry. Exosomes released by LnCap prostate cancer cells were also purified using a modified procedure based on polyethylene glycol (PEG) precipitation. Results: Exosomes extracted from LnCap cells contained higher proportions of phosphatidyl choline, phosphatidyl ethanolamine and phosphatidyl inositol lipids than whole LnCap cells. Lysophosphatidylcholine, a harmful intermediate product of phosphatidylcholine metabolism in vivo, was not found in LnCap cells but in exosomes. Phospholipids were different in exosomes from LnCap, PC3 and DU-145 prostate cancer cells. The main lipid pathways involved, i.e., glycerophospholipid metabolism, autophagy, and ferroptosis pathways, were also different in these cells. Exosomes isolated by this modified PEG precipitation technique were similar in purity to those obtained using a commercial kit. Conclusions: This study demonstrates that phosphatidylcholine and its harmful product lysophosphatidylcholine may play important roles in hormone-sensitive prostate cancer. Phospholipid exosome metabolism was changed in hormone-sensitive and hormone-resistant prostate cancer cells. The LPC, lipid pathway of autophagy and ferroptosis may act as therapeutic targets. The possibility of purifying prostate cancer cell exosomes using modified PEG precipitation is suitable for cancer screening.
Collapse
Affiliation(s)
- Xianlin Yi
- Department of Urology, The Affiliated Cancer Hospital of Guangxi Medical University & Guangxi Cancer Research Institute, Nanning 530021,China
| | - You Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China.,Life science institute of East China Normal University, Shanghai 200241, P.R. China
| | - XiaoGang Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - FuBing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Tiangang Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China.,Wuhan infectious diseases and cancer research center, Chinese Academy of Medical Sciences, Wuhan 430071, P.R. China.,Hubei Engineering Laboratory for Synthetic Microbiology, Wuhan Institute of Biotechnology, Wuhan 430075, PR China
| |
Collapse
|
21
|
Devkota R, Henricsson M, Borén J, Pilon M. The C. elegans PAQR-2 and IGLR-2 membrane homeostasis proteins are uniquely essential for tolerating dietary saturated fats. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158883. [PMID: 33444761 DOI: 10.1016/j.bbalip.2021.158883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 01/07/2023]
Abstract
How cells maintain vital membrane lipid homeostasis while obtaining most of their constituent fatty acids from a varied diet remains largely unknown. Here, we report the first whole-organism (Caenorhabditis elegans) forward genetic screen to identify genes essential for tolerance to dietary saturated fatty acids (SFAs). We found that only the PAQR-2/IGLR-2 pathway, homologous to the human adiponectin receptor 2 (AdipoR2) pathway, is uniquely essential to prevent SFA-mediated toxicity. When provided a SFA-rich diet, worms lacking either protein accumulate an excess of SFAs in their membrane phospholipids, which is accompanied by membrane rigidification. Additionally, we used fluorescence resonance energy transfer (FRET) to show that the interaction between PAQR-2 and IGLR-2 is regulated by membrane fluidity, suggesting a mechanism by which this protein complex senses membrane properties. We also created versions of PAQR-2 that lacked parts of the cytoplasmic N-terminal domain and showed that these were still functional, though still dependent on the interaction with IGLR-2. We conclude that membrane homeostasis via the PAQR-2/IGLR-2 fluidity sensor is the only pathway specifically essential for the non-toxic uptake of dietary SFAs in C. elegans.
Collapse
Affiliation(s)
- Ranjan Devkota
- Dept. Chemistry and Molecular Biology, Univ. Gothenburg, 405 30 Gothenburg, Sweden
| | - Marcus Henricsson
- Dept. Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30 Gothenburg, Sweden
| | - Jan Borén
- Dept. Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30 Gothenburg, Sweden
| | - Marc Pilon
- Dept. Chemistry and Molecular Biology, Univ. Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
22
|
Ruiz M, Palmgren H, Henricsson M, Devkota R, Jaiswal H, Maresca M, Bohlooly-Y M, Peng XR, Borén J, Pilon M. Extensive transcription mis-regulation and membrane defects in AdipoR2-deficient cells challenged with saturated fatty acids. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158884. [PMID: 33444759 DOI: 10.1016/j.bbalip.2021.158884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022]
Abstract
How cells maintain vital membrane lipid homeostasis while obtaining most of their constituent fatty acids from a varied diet remains largely unknown. Here, we used transcriptomics, lipidomics, growth and respiration assays, and membrane property analyses in human HEK293 cells or human umbilical vein endothelial cells (HUVEC) to show that the function of AdipoR2 is to respond to membrane rigidification by regulating many lipid metabolism genes. We also show that AdipoR2-dependent membrane homeostasis is critical for growth and respiration in cells challenged with saturated fatty acids. Additionally, we found that AdipoR2 deficiency causes transcriptome and cell physiological defects similar to those observed in SREBP-deficient cells upon SFA challenge. Finally, we compared several genes considered important for lipid homeostasis, namely AdipoR2, SCD, FADS2, PEMT and ACSL4, and found that AdipoR2 and SCD are the most important among these to prevent membrane rigidification and excess saturation when human cells are challenged with exogenous SFAs. We conclude that AdipoR2-dependent membrane homeostasis is one of the primary mechanisms that protects against exogenous SFAs.
Collapse
Affiliation(s)
- Mario Ruiz
- Dept. Chemistry and Molecular Biology, Univ. Gothenburg, 405 30 Gothenburg, Sweden
| | - Henrik Palmgren
- Metabolism Bioscience, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marcus Henricsson
- Dept. Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ranjan Devkota
- Dept. Chemistry and Molecular Biology, Univ. Gothenburg, 405 30 Gothenburg, Sweden
| | - Himjyot Jaiswal
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden; CellinkAB, Arvid Wallgrens Backe 20, 413 46 Gothenburg, Sweden
| | - Marcello Maresca
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Xiao-Rong Peng
- Metabolism Bioscience, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jan Borén
- Dept. Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Univ. of Gothenburg, 405 30 Gothenburg, Sweden
| | - Marc Pilon
- Dept. Chemistry and Molecular Biology, Univ. Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
23
|
Beaufrère H, Gardhouse SM, Wood RD, Stark KD. The plasma lipidome of the Quaker parrot (Myiopsitta monachus). PLoS One 2020; 15:e0240449. [PMID: 33259543 PMCID: PMC7707497 DOI: 10.1371/journal.pone.0240449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/25/2020] [Indexed: 11/19/2022] Open
Abstract
Dyslipidemias and lipid-accumulation disorders are common in captive parrots, in particular in Quaker parrots. Currently available diagnostic tests only measure a fraction of blood lipids and have overall problematic cross-species applicability. Comprehensively analyzing lipids in the plasma of parrots is the first step to better understand their lipid metabolism in health and disease, as well as to explore new lipid biomarkers. The plasma lipidome of 12 Quaker parrots was investigated using UHPLC-MS/MS with both targeted and untargeted methods. Targeted methods on 6 replicates measured 432 lipids comprised of sterol, cholesterol ester, bile acid, fatty acid, acylcarnitine, glycerolipid, glycerophospholipid, and sphingolipid panels. For untargeted lipidomics, precursor ion mass-to-charge ratios were matched to corresponding lipids using the LIPIDMAPS structure database and LipidBlast at the sum composition or acyl species level of information. Sterol lipids and glycerophospholipids constituted the majority of plasma lipids on a molar basis. The most common lipids detected with the targeted methods included free cholesterol, CE(18:2), CE(20:4) for sterol lipids; PC(36:2), PC(34:2), PC(34:1) for glycerophospholipids; TG(52:3), TG(54:4), TG(54:5), TG(52:2) for glycerolipids; SM(d18:1/16:0) for sphingolipids; and palmitic acid for fatty acyls. Over a thousand different lipid species were detected by untargeted lipidomics. Sex differences in the plasma lipidome were observed using heatmaps, principal component analysis, and discriminant analysis. This report presents the first comprehensive database of plasma lipid species in psittacine birds and paves the way for further research into blood lipid diagnostics and the impact of diet, diseases, and drugs on the parrot plasma lipidome.
Collapse
Affiliation(s)
- Hugues Beaufrère
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
- * E-mail:
| | - Sara M. Gardhouse
- Health Sciences Centre, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - R. Darren Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ken D. Stark
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
24
|
Zardini Buzatto A, Kwon BK, Li L. Development of a NanoLC-MS workflow for high-sensitivity global lipidomic analysis. Anal Chim Acta 2020; 1139:88-99. [DOI: 10.1016/j.aca.2020.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022]
|
25
|
Zelnik ID, Volpert G, Viiri LE, Kauhanen D, Arazi T, Aalto-Setälä K, Laaksonen R, Futerman AH. Different rates of flux through the biosynthetic pathway for long-chain versus very-long-chain sphingolipids. J Lipid Res 2020; 61:1341-1346. [PMID: 32651186 PMCID: PMC7529049 DOI: 10.1194/jlr.ra120000984] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The backbone of all sphingolipids (SLs) is a sphingoid long-chain base (LCB) to which a fatty acid is N-acylated. Considerable variability exists in the chain length and degree of saturation of both of these hydrophobic chains, and recent work has implicated ceramides with different LCBs and N-acyl chains in distinct biological processes; moreover, they may play different roles in disease states and possibly even act as prognostic markers. We now demonstrate that the half-life, or turnover rate, of ceramides containing diverse N-acyl chains is different. By means of a pulse-labeling protocol using stable-isotope, deuterated free fatty acids, and following their incorporation into ceramide and downstream SLs, we show that very-long-chain (VLC) ceramides containing C24:0 or C24:1 fatty acids turn over much more rapidly than long-chain (LC) ceramides containing C16:0 or C18:0 fatty acids due to the more rapid metabolism of the former into VLC sphingomyelin and VLC hexosylceramide. In contrast, d16:1 and d18:1 ceramides show similar rates of turnover, indicating that the length of the sphingoid LCB does not influence the flux of ceramides through the biosynthetic pathway. Together, these data demonstrate that the N-acyl chain length of SLs may not only affect membrane biophysical properties but also influence the rate of metabolism of SLs so as to regulate their levels and perhaps their biological functions.
Collapse
Affiliation(s)
- Iris D Zelnik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Giora Volpert
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Leena E Viiri
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere Finland
| | | | - Tamar Arazi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Katriina Aalto-Setälä
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere Finland
| | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
26
|
Miniewska K, Godzien J, Mojsak P, Maliszewska K, Kretowski A, Ciborowski M. Mass spectrometry-based determination of lipids and small molecules composing adipose tissue with a focus on brown adipose tissue. J Pharm Biomed Anal 2020; 191:113623. [PMID: 32966938 DOI: 10.1016/j.jpba.2020.113623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
Adipose tissue has been the subject of research for a very long time. Many studies perform a comprehensive analysis of different types of adipose tissue with an emphasis on brown adipose tissue. Mass spectrometry-based approaches are particularly useful in the exploration not only of the metabolic composition of adipose tissue but also its function. In the presented review, a complex and critical overview of publications devoted to the analysis of adipose tissue by means of mass spectrometry was performed. Detailed investigation of analytical aspects related to either untargeted or targeted analysis of adipose tissue was performed, leading to the formation of a collection of hints at the available analytical methods. Moreover, a profound analysis of the metabolic composition of brown adipose tissue was performed. Brown adipose tissue metabolome was characterized on structural and functional levels, providing information about its exact metabolic composition but also connecting these molecules and placing them into biochemical pathways. All our work resulted in a very broad picture of the analysis of adipose tissue, starting from the analytical aspects and finishing on the current knowledge about its composition.
Collapse
Affiliation(s)
- Katarzyna Miniewska
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Godzien
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Patrycja Mojsak
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
27
|
Beyene HB, Olshansky G, T. Smith AA, Giles C, Huynh K, Cinel M, Mellett NA, Cadby G, Hung J, Hui J, Beilby J, Watts GF, Shaw JS, Moses EK, Magliano DJ, Meikle PJ. High-coverage plasma lipidomics reveals novel sex-specific lipidomic fingerprints of age and BMI: Evidence from two large population cohort studies. PLoS Biol 2020; 18:e3000870. [PMID: 32986697 PMCID: PMC7544135 DOI: 10.1371/journal.pbio.3000870] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/08/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity and related metabolic diseases show clear sex-related differences. The growing burden of these diseases calls for better understanding of the age- and sex-related metabolic consequences. High-throughput lipidomic analyses of population-based cohorts offer an opportunity to identify disease-risk-associated biomarkers and to improve our understanding of lipid metabolism and biology at a population level. Here, we comprehensively examined the relationship between lipid classes/subclasses and molecular species with age, sex, and body mass index (BMI). Furthermore, we evaluated sex specificity in the association of the plasma lipidome with age and BMI. Some 747 targeted lipid measures, representing 706 molecular lipid species across 36 classes/subclasses, were measured using a high-performance liquid chromatography coupled mass spectrometer on a total of 10,339 participants from the Australian Diabetes, Obesity and Lifestyle Study (AusDiab), with 563 lipid species being validated externally on 4,207 participants of the Busselton Health Study (BHS). Heat maps were constructed to visualise the relative differences in lipidomic profile between men and women. Multivariable linear regression analyses, including sex-interaction terms, were performed to assess the associations of lipid species with cardiometabolic phenotypes. Associations with age and sex were found for 472 (66.9%) and 583 (82.6%) lipid species, respectively. We further demonstrated that age-associated lipidomic fingerprints differed by sex. Specific classes of ether-phospholipids and lysophospholipids (calculated as the sum composition of the species within the class) were inversely associated with age in men only. In analyses with women alone, higher triacylglycerol and lower lysoalkylphosphatidylcholine species were observed among postmenopausal women compared with premenopausal women. We also identified sex-specific associations of lipid species with obesity. Lysophospholipids were negatively associated with BMI in both sexes (with a larger effect size in men), whilst acylcarnitine species showed opposing associations based on sex (positive association in women and negative association in men). Finally, by utilising specific lipid ratios as a proxy for enzymatic activity, we identified stearoyl CoA desaturase (SCD-1), fatty acid desaturase 3 (FADS3), and plasmanylethanolamine Δ1-desaturase activities, as well as the sphingolipid metabolic pathway, as constituent perturbations of cardiometabolic phenotypes. Our analyses elucidate the effect of age and sex on lipid metabolism by offering a comprehensive view of the lipidomic profiles associated with common cardiometabolic risk factors. These findings have implications for age- and sex-dependent lipid metabolism in health and disease and suggest the need for sex stratification during lipid biomarker discovery, establishing biological reference intervals for assessment of disease risk.
Collapse
Affiliation(s)
- Habtamu B. Beyene
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | | | | | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | | | - Gemma Cadby
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Joseph Hung
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Jennie Hui
- School of Population and Global Health, University of Western Australia, Perth, Australia
- PathWest Laboratory Medicine of Western Australia, Nedlands, Western Australia
| | - John Beilby
- PathWest Laboratory Medicine of Western Australia, Nedlands, Western Australia
| | - Gerald F. Watts
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | | | - Eric K. Moses
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Dianna J. Magliano
- Baker Heart and Diabetes Institute, Melbourne, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
28
|
Shen M, Di K, He H, Xia Y, Xie H, Huang R, Liu C, Yang M, Zheng S, He N, Li Z. Progress in exosome associated tumor markers and their detection methods. MOLECULAR BIOMEDICINE 2020; 1:3. [PMID: 35006428 PMCID: PMC8603992 DOI: 10.1186/s43556-020-00002-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Exosomes are secreted by cells and are widely present in body fluids. Exosomes contain various molecular constituents of their cells of origin such as proteins, mRNA, miRNAs, DNA, lipid and glycans which are very similar as the content in tumor cells. These contents play an important role in various stages of tumor development, and make the tumor-derived exosome as a hot and emerging biomarker for various cancers diagnosis and management in non-invasive manner. The present problems of exosome isolation and detection hinder the application of exosomes. With the development of exosome isolation and detection technology, the contents of exosomes can be exploited for early cancer diagnosis. This review summarizes the recent progress on exosome-associated tumor biomarkers and some new technologies for exosome isolation and detection. Furthermore, we have also discussed the future development direction in exosome analysis methods.
Collapse
Affiliation(s)
- Mengjiao Shen
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Shanghai Health Development Research Center, Shanghai, China
| | - Kaili Di
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hongzhang He
- Captis Diagnostics Inc, Pittsburgh, PA, 15213, USA
| | - Yanyan Xia
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hui Xie
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Rongrong Huang
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chang Liu
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mo Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong, People's Republic of China.
| | - Siyang Zheng
- Department of Biomedical Engineering and Electrical & Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Scott Hall 4N211, Pittsburgh, PA, 15213, USA.
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
29
|
Leveraging a gain-of-function allele of Caenorhabditis elegans paqr-1 to elucidate membrane homeostasis by PAQR proteins. PLoS Genet 2020; 16:e1008975. [PMID: 32750056 PMCID: PMC7428288 DOI: 10.1371/journal.pgen.1008975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/14/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
The C. elegans proteins PAQR-2 (a homolog of the human seven-transmembrane domain AdipoR1 and AdipoR2 proteins) and IGLR-2 (a homolog of the mammalian LRIG proteins characterized by a single transmembrane domain and the presence of immunoglobulin domains and leucine-rich repeats in their extracellular portion) form a complex that protects against plasma membrane rigidification by promoting the expression of fatty acid desaturases and the incorporation of polyunsaturated fatty acids into phospholipids, hence increasing membrane fluidity. In the present study, we leveraged a novel gain-of-function allele of PAQR-1, a PAQR-2 paralog, to carry out structure-function studies. We found that the transmembrane domains of PAQR-2 are responsible for its functional requirement for IGLR-2, that PAQR-1 does not require IGLR-2 but acts via the same pathway as PAQR-2, and that the divergent N-terminal cytoplasmic domains of the PAQR-1 and PAQR-2 proteins serve a regulatory function and may regulate access to the catalytic site of these proteins. We also show that overexpression of human AdipoR1 or AdipoR2 alone is sufficient to confer increased palmitic acid resistance in HEK293 cells, and thus act in a manner analogous to the PAQR-1 gain-of-function allele.
Collapse
|
30
|
Ekroos K, Lavrynenko O, Titz B, Pater C, Hoeng J, Ivanov NV. Lipid-based biomarkers for CVD, COPD, and aging - A translational perspective. Prog Lipid Res 2020; 78:101030. [PMID: 32339553 DOI: 10.1016/j.plipres.2020.101030] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/23/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
For many diseases, there is an unmet need for new or better biomarkers for improved disease risk assessment and monitoring, as available markers lack sufficient specificity. Lipids are drawing major interest as potential candidates for filling these gaps. This has recently been demonstrated by the identification of selective ceramides for prediction of cardiovascular mortality, enabling improved risk assessment of cardiovascular disease compared with conventional clinical markers. In this review, we discuss current lipid biomarker findings and the possible connection between cardiovascular disease, chronic obstructive pulmonary disease, and aging. Moreover, we discuss how to overcome the current roadblocks facing lipid biomarker research. We stress the need for improved quantification, standardization of methodologies, and establishment of initial reference values to allow for an efficient transfer path of research findings into the clinical landscape, and, ultimately, to put newly identified biomarkers into practical use.
Collapse
Affiliation(s)
- Kim Ekroos
- Lipidomics Consulting Ltd., Irisviksvägen 31D, 02230 Esbo, Finland.
| | - Oksana Lavrynenko
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Calin Pater
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchâtel, Switzerland.
| |
Collapse
|
31
|
Skotland T, Sagini K, Sandvig K, Llorente A. An emerging focus on lipids in extracellular vesicles. Adv Drug Deliv Rev 2020; 159:308-321. [PMID: 32151658 DOI: 10.1016/j.addr.2020.03.002] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/02/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles contain a lipid bilayer membrane that protects the encapsulated material, such as proteins, nucleic acids, lipids and metabolites, from the extracellular environment. These vesicles are released from cells via different mechanisms. During recent years extracellular vesicles have been studied as possible biomarkers for different diseases, as biological nanoparticles for drug delivery, and in basic studies as a tool to understand the structure of biological membranes and the mechanisms involved in vesicular trafficking. Lipids are essential molecular components of extracellular vesicles, but at the moment our knowledge about the lipid composition and the function of lipids in these vesicles is limited. However, the interest of the research community in these molecules is increasing as their role in extracellular vesicles is starting to be acknowledged. In this review, we will present the status of the field and describe what is needed to bring it forward.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway; Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway.
| |
Collapse
|
32
|
Wang J, Han X. Analytical challenges of shotgun lipidomics at different resolution of measurements. Trends Analyt Chem 2019; 121:115697. [PMID: 32713986 PMCID: PMC7382544 DOI: 10.1016/j.trac.2019.115697] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The essence of shotgun lipidomics is to maintain consistency of the chemical environment of lipid samples during mass spectrometry acquisition. This strategy is suitable for large-scale quantitative analysis. This strategy also allows sufficient time to collect data to improve the signal-to-noise ratio. The initial approach of shotgun lipidomics was the electrospray ionization (ESI)-based direct infusion mass spectrometry strategy. With development of mass spectrometry for small molecules, shotgun lipidomics methods have been extended to matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS) and ambient mass spectrometry, including MS imaging methods. Furthermore, the object of analysis has extended from organ and body fluid levels to tissue and cell levels with technological developments. In this article, we summarize the status and technical challenges of shotgun lipidomics at different resolution of measurements from the mass spectrometry perspective.
Collapse
Affiliation(s)
- Jianing Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
- Department of Medicine – Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
33
|
Ruiz M, Bodhicharla R, Ståhlman M, Svensk E, Busayavalasa K, Palmgren H, Ruhanen H, Boren J, Pilon M. Evolutionarily conserved long-chain Acyl-CoA synthetases regulate membrane composition and fluidity. eLife 2019; 8:47733. [PMID: 31769755 PMCID: PMC6901333 DOI: 10.7554/elife.47733] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/23/2019] [Indexed: 12/19/2022] Open
Abstract
The human AdipoR1 and AdipoR2 proteins, as well as their C. elegans homolog PAQR-2, protect against cell membrane rigidification by exogenous saturated fatty acids by regulating phospholipid composition. Here, we show that mutations in the C. elegans gene acs-13 help to suppress the phenotypes of paqr-2 mutant worms, including their characteristic membrane fluidity defects. acs-13 encodes a homolog of the human acyl-CoA synthetase ACSL1, and localizes to the mitochondrial membrane where it likely activates long chains fatty acids for import and degradation. Using siRNA combined with lipidomics and membrane fluidity assays (FRAP and Laurdan dye staining) we further show that the human ACSL1 potentiates lipotoxicity by the saturated fatty acid palmitate: silencing ACSL1 protects against the membrane rigidifying effects of palmitate and acts as a suppressor of AdipoR2 knockdown, thus echoing the C. elegans findings. We conclude that acs-13 mutations in C. elegans and ACSL1 knockdown in human cells prevent lipotoxicity by promoting increased levels of polyunsaturated fatty acid-containing phospholipids.
Collapse
Affiliation(s)
- Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Rakesh Bodhicharla
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Emma Svensk
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Kiran Busayavalasa
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Palmgren
- Metabolism BioScience, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science, Helsinki, Finland.,Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Jan Boren
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Wang J, Wang C, Han X. Tutorial on lipidomics. Anal Chim Acta 2019; 1061:28-41. [PMID: 30926037 PMCID: PMC7375172 DOI: 10.1016/j.aca.2019.01.043] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
The mainstream of lipidomics involves mass spectrometry-based, systematic, and large-scale studies of the structure, composition, and quantity of lipids in biological systems such as organs, cells, and body fluids. As increasingly more researchers in broad fields are beginning to pay attention to and actively learn about the lipidomic technology, some introduction on the topic is needed to help the newcomers to better understand the field. This tutorial seeks to introduce the basic knowledge about lipidomics and to provide readers with some core ideas and the most important approaches for studying the field.
Collapse
Affiliation(s)
- Jianing Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Chunyan Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Medicine - Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
35
|
Skotland T, Sandvig K. The role of PS 18:0/18:1 in membrane function. Nat Commun 2019; 10:2752. [PMID: 31227693 PMCID: PMC6588574 DOI: 10.1038/s41467-019-10711-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
Various studies have demonstrated that the two leaflets of cellular membranes interact, potentially through so-called interdigitation between the fatty acyl groups. While the molecular mechanism underlying interleaflet coupling remains to be fully understood, recent results suggest interactions between the very-long-chain sphingolipids in the outer leaflet, and phosphatidylserine PS18:0/18:1 in the inner leaflet, and an important role for cholesterol for these interactions. Here we review the evidence that cross-linking of sphingolipids may result in clustering of phosphatidylserine and transfer of signals to the cytosol. Although much remains to be uncovered, the molecular properties and abundance of PS 18:0/18:1 suggest a unique role for this lipid. There are several lines of evidence for interactions between the two membrane leaflets in cells. In this review the authors discuss the transmembrane coupling of lipids, the involvement of phosphatidyl serine species PS 18:0/18:1, and their importance for various cellular processes.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.,Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
36
|
Fumeron F, Nicolas A, Bastard JP, Fellahi S, Wigger L, Ibberson M, Cruciani-Guglielmacci C, Le Stunff H, Velho G, Magnan C, Marre M, Balkau B, Roussel R. Dairy consumption is associated with lower plasma dihydroceramides in women from the D.E.S.I.R. cohort. DIABETES & METABOLISM 2019; 46:144-149. [PMID: 31212089 DOI: 10.1016/j.diabet.2019.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/15/2019] [Accepted: 06/09/2019] [Indexed: 12/20/2022]
Abstract
AIM In the D.E.S.I.R. cohort, higher consumption of dairy products was associated with lower incidence of hyperglycaemia, and dihydroceramide concentrations were higher in those who progressed to diabetes. Our aim here was to study the relationships between dairy consumption and concentrations of dihydroceramides and ceramides. METHODS In the D.E.S.I.R. cohort, men and women aged 30-65 years, volunteers from West-Central France, were included in a 9-year follow-up with examinations every 3 years, including food-frequency questionnaires. Two items concerned dairy products (cheese, other dairy products except cheese). At each examination, dihydroceramides and ceramides were determined by mass spectrometry in a cohort subset; in the present study, the 105 people who did not progress to type 2 diabetes were analyzed, as the disorder per se might be a confounding factor. RESULTS Higher consumption of dairy products (except cheese) was associated with total plasma dihydroceramides during the follow-up, but only in women (P=0.01 for gender interaction). In fact, dihydroceramide levels were lower in women with high vs low consumption (P=0.03), and were significantly increased during follow-up (P=0.01) in low consumers only. There was also a trend for lower ceramides in women with high dairy (except cheese) intakes (P=0.08). Cheese was associated with dihydroceramide and ceramide changes during follow-up (P=0.04 for both), but no clear trend was evident in either low or high consumers. CONCLUSION These results show that, in women, there is an inverse association between fresh dairy product consumption and predictive markers (dihydroceramides) of type 2 diabetes.
Collapse
Affiliation(s)
- F Fumeron
- Centre de Recherche des Cordeliers, Inserm UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Université de Paris, UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France.
| | - A Nicolas
- Centre de Recherche des Cordeliers, Inserm UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Sorbonne Université, 15 rue de l'école de Médecine, 75006 Paris, France
| | - J-P Bastard
- AP-HP, Biochemistry and Hormonology Department, Tenon Hospital, 4 Rue de la Chine, 75020 Paris, France
| | - S Fellahi
- AP-HP, Biochemistry and Hormonology Department, Tenon Hospital, 4 Rue de la Chine, 75020 Paris, France
| | - L Wigger
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - M Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - C Cruciani-Guglielmacci
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université de Paris, 4 rue Marie-Andrée Lagroua Weill Hallé, 75013 Paris, France
| | - H Le Stunff
- UMR 9198 Institut des Neurosciences Paris Saclay (Neuro-PSI), Université Paris-Sud, Université Paris-Saclay, bâtiment 447, 91405 Orsay cedex, France
| | - G Velho
- Centre de Recherche des Cordeliers, Inserm UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France
| | - C Magnan
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université de Paris, 4 rue Marie-Andrée Lagroua Weill Hallé, 75013 Paris, France
| | - M Marre
- Centre de Recherche des Cordeliers, Inserm UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Université de Paris, UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Diabetology, Endocrinology, Nutrition, APHP - Bichat Hospital, 46 rue Henri Huchard, 75018 Paris, France
| | - B Balkau
- Centre for Research in Epidemiology and Population Health (CESP), INSERM, UMR-S 1018, University Paris-Sud, University Versailles Saint-Quentin, 16 av. Paul Vaillant Couturier 94800 Villejuif, France
| | - R Roussel
- Centre de Recherche des Cordeliers, Inserm UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Université de Paris, UMR-S 1138, 15 rue de l'école de Médecine, 75006 Paris, France; Diabetology, Endocrinology, Nutrition, APHP - Bichat Hospital, 46 rue Henri Huchard, 75018 Paris, France
| |
Collapse
|
37
|
Takeda H, Takahashi M, Hara T, Izumi Y, Bamba T. Improved quantitation of lipid classes using supercritical fluid chromatography with a charged aerosol detector. J Lipid Res 2019; 60:1465-1474. [PMID: 31201290 DOI: 10.1194/jlr.d094516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/11/2019] [Indexed: 01/01/2023] Open
Abstract
Quantitatively and rapidly analyzing lipids is necessary to elucidate their biological functions. Herein, we developed a quantitative method for various lipid classes using supercritical fluid chromatography (SFC) coupled with a charged aerosol detector (CAD), providing high-throughput data analysis to detect a large number of molecules in each lipid class as one peak. Applying the CAD was useful for analyzing lipid molecules in the same lipid class with a constant response under the same mobile phase composition. First, we optimized the washing method for the diethylamine column, achieving baseline separation of lipid classes while maintaining good peak shapes. In addition, the CAD conditions (organic solvent evaporation and numerical correction of the CAD data) were optimized to improve the signal-to-noise ratio. We used an internal standard (ceramide phosphoethanolamine d17:1-12:0), which did not coelute with the lipid classes and showed high extraction efficiency. Based on a quantitative analysis of HepG2 cells, the concentration of lipid classes detected by CAD was adequate compared with that obtained by triple-quadrupole MS (QqQMS) in a previous study because the deviations of the concentrations were 0.6- to 2.3-fold. These results also supported the quantitative performance of SFC-QqQMS developed in our previous report.
Collapse
Affiliation(s)
- Hiroaki Takeda
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Hara
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
38
|
Ruiz M, Ståhlman M, Borén J, Pilon M. AdipoR1 and AdipoR2 maintain membrane fluidity in most human cell types and independently of adiponectin. J Lipid Res 2019; 60:995-1004. [PMID: 30890562 PMCID: PMC6495173 DOI: 10.1194/jlr.m092494] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/07/2019] [Indexed: 12/16/2022] Open
Abstract
The FA composition of phospholipids must be tightly regulated to maintain optimal cell membrane properties and compensate for a highly variable supply of dietary FAs. Previous studies have shown that AdipoR2 and its homologue PAQR-2 are important regulators of phospholipid FA composition in HEK293 cells and Caenorhabditiselegans, respectively. Here we show that both AdipoR1 and AdipoR2 are essential for sustaining desaturase expression and high levels of unsaturated FAs in membrane phospholipids of many human cell types, including primary human umbilical vein endothelial cells, and for preventing membrane rigidification in cells challenged with exogenous palmitate, a saturated FA. Three independent methods confirm the role of the AdipoRs as regulators of membrane composition and fluidity: fluorescence recovery after photobleaching, measurements of Laurdan dye generalized polarization, and mass spectrometry to determine the FA composition of phospholipids. Furthermore, we show that the AdipoRs can prevent lipotoxicity in the complete absence of adiponectin, their putative ligand. We propose that the primary cellular function of AdipoR1 and AdipoR2 is to maintain membrane fluidity in most human cell types and that adiponectin is not required for this function.
Collapse
Affiliation(s)
- Mario Ruiz
- Department of Chemistry and Molecular Biology; University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology; University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
39
|
Ruiz M, Bodhicharla R, Svensk E, Devkota R, Busayavalasa K, Palmgren H, Ståhlman M, Boren J, Pilon M. Membrane fluidity is regulated by the C. elegans transmembrane protein FLD-1 and its human homologs TLCD1/2. eLife 2018; 7:e40686. [PMID: 30509349 PMCID: PMC6279351 DOI: 10.7554/elife.40686] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/04/2018] [Indexed: 12/15/2022] Open
Abstract
Dietary fatty acids are the main building blocks for cell membranes in animals, and mechanisms must therefore exist that compensate for dietary variations. We isolated C. elegans mutants that improved tolerance to dietary saturated fat in a sensitized genetic background, including eight alleles of the novel gene fld-1 that encodes a homolog of the human TLCD1 and TLCD2 transmembrane proteins. FLD-1 is localized on plasma membranes and acts by limiting the levels of highly membrane-fluidizing long-chain polyunsaturated fatty acid-containing phospholipids. Human TLCD1/2 also regulate membrane fluidity by limiting the levels of polyunsaturated fatty acid-containing membrane phospholipids. FLD-1 and TLCD1/2 do not regulate the synthesis of long-chain polyunsaturated fatty acids but rather limit their incorporation into phospholipids. We conclude that inhibition of FLD-1 or TLCD1/2 prevents lipotoxicity by allowing increased levels of membrane phospholipids that contain fluidizing long-chain polyunsaturated fatty acids. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Mario Ruiz
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | - Rakesh Bodhicharla
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | - Emma Svensk
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | - Ranjan Devkota
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | - Kiran Busayavalasa
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| | - Henrik Palmgren
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
- Diabetes Bioscience, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of MedicineUniversity of GothenburgGothenburgSweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of MedicineUniversity of GothenburgGothenburgSweden
| | - Marc Pilon
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
40
|
Gathungu RM, Larrea P, Sniatynski MJ, Marur VR, Bowden JA, Koelmel JP, Starke-Reed P, Hubbard VS, Kristal BS. Optimization of Electrospray Ionization Source Parameters for Lipidomics To Reduce Misannotation of In-Source Fragments as Precursor Ions. Anal Chem 2018; 90:13523-13532. [PMID: 30265528 PMCID: PMC6297073 DOI: 10.1021/acs.analchem.8b03436] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lipidomics requires the accurate annotation of lipids in complex samples to enable determination of their biological relevance. We demonstrate that unintentional in-source fragmentation (ISF, common in lipidomics) generates ions that have identical masses to other lipids. Lysophosphatidylcholines (LPC), for example, generate in-source fragments with the same mass as free fatty acids and lysophosphatidylethanolamines (LPE). The misannotation of in-source fragments as true lipids is particularly insidious in complex matrixes since most masses are initially unannotated and comprehensive lipid standards are unavailable. Indeed, we show such LPE/LPC misannotations are incorporated in the data submitted to the National Institute of Standards and Technology (NIST) interlaboratory comparison exercise. Computer simulations exhaustively identified potential misannotations. The selection of in-source fragments of highly abundant lipids as features, instead of the correct recognition of trace lipids, can potentially lead to (i) missing the biologically relevant lipids (i.e., a false negative) and/or (ii) incorrect assignation of a phenotype to an incorrect lipid (i.e., false positive). When ISF is not eliminated in the negative ion mode, ∼40% of the 100 most abundant masses corresponding to unique phospholipids measured in plasma were artifacts from ISF. We show that chromatographic separation and ion intensity considerations assist in distinguishing precursor ions from in-source fragments, suggesting ISF may be especially problematic when complex samples are analyzed via shotgun lipidomics. We also conduct a systematic evaluation of electrospray ionization (ESI) source parameters on an Exactive equipped with a heated electrospray ionization (HESI-II) source with the objective of obtaining uniformly appropriate source conditions for a wide range of lipids, while, at the same time, reducing in-source fragmentation.
Collapse
Affiliation(s)
- Rose M. Gathungu
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Department of Medicine, Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| | - Pablo Larrea
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Department of Medicine, Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| | - Matthew J. Sniatynski
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Department of Medicine, Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| | - Vasant R. Marur
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Department of Medicine, Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| | - John A. Bowden
- Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610
- National Institute of Standards and Technology, Hollings Marine Laboratory, Charleston, SC 29412
| | - Jeremy P. Koelmel
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Pamela Starke-Reed
- Deputy Director, NIH Division of Nutrition Research Coordination, Bethesda, MD 20892
| | - Van S. Hubbard
- Director, NIH Division of Nutrition Research Coordination, Bethesda, MD 20892
| | - Bruce S. Kristal
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Department of Medicine, Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
41
|
Burla B, Arita M, Arita M, Bendt AK, Cazenave-Gassiot A, Dennis EA, Ekroos K, Han X, Ikeda K, Liebisch G, Lin MK, Loh TP, Meikle PJ, Orešič M, Quehenberger O, Shevchenko A, Torta F, Wakelam MJO, Wheelock CE, Wenk MR. MS-based lipidomics of human blood plasma: a community-initiated position paper to develop accepted guidelines. J Lipid Res 2018; 59:2001-2017. [PMID: 30115755 PMCID: PMC6168311 DOI: 10.1194/jlr.s087163] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/11/2018] [Indexed: 12/19/2022] Open
Abstract
Human blood is a self-regenerating lipid-rich biological fluid that is routinely collected in hospital settings. The inventory of lipid molecules found in blood plasma (plasma lipidome) offers insights into individual metabolism and physiology in health and disease. Disturbances in the plasma lipidome also occur in conditions that are not directly linked to lipid metabolism; therefore, plasma lipidomics based on MS is an emerging tool in an array of clinical diagnostics and disease management. However, challenges exist in the translation of such lipidomic data to clinical applications. These relate to the reproducibility, accuracy, and precision of lipid quantitation, study design, sample handling, and data sharing. This position paper emerged from a workshop that initiated a community-led process to elaborate and define a set of generally accepted guidelines for quantitative MS-based lipidomics of blood plasma or serum, with harmonization of data acquired on different instrumentation platforms across independent laboratories as an ultimate goal. We hope that other fields may benefit from and follow such a precedent.
Collapse
Affiliation(s)
- Bo Burla
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Masanori Arita
- National Institute of Genetics, Shizuoka, Japan and RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | - Anne K Bendt
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore
| | - Edward A Dennis
- Departments of Pharmacology and Chemistry and Biochemistry, School of Medicine, University of California at San Diego, La Jolla, CA
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Esbo, Finland
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies and Department of Medicine-Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Michelle K Lin
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, Singapore
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland and School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Oswald Quehenberger
- Departments of Pharmacology and Medicine, School of Medicine, University of California at San Diego, La Jolla, CA
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Federico Torta
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore
| | | | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
42
|
Wang C, Wang C, Liu F, Rainosek S, Patterson TA, Slikker W, Han X. Lipidomics Reveals Changes in Metabolism, Indicative of Anesthetic-Induced Neurotoxicity in Developing Brains. Chem Res Toxicol 2018; 31:825-835. [PMID: 30132657 DOI: 10.1021/acs.chemrestox.8b00186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Numerous studies have demonstrated that treatment with high dose anesthetics for a prolonged duration induces brain injury in infants. However, whether anesthetic treatment leading to neurotoxicity is associated with alterations in lipid metabolism and homeostasis is still unclear. This review first outlines the lipidomics tools for analysis of lipid molecular species that can inform alterations in lipid species after anesthetic exposure. Then the available data indicating anesthetics cause changes in lipid profiles in the brain and serum of infant monkeys in preclinical studies are summarized, and the potential mechanisms leading to the altered lipid metabolism and their association with anesthetic-induced brain injury are also discussed. Finally, whether lipid changes identified in serum of infant monkeys can serve as indicators for the early detection of anesthetic-induced brain injury is described. We believe extensive studies on alterations in lipids after exposure to anesthetics will allow us to better understand anesthetic-induced neurotoxicity, unravel its underlying biochemical mechanisms, and develop powerful biomarkers for early detection/monitoring of the toxicity.
Collapse
Affiliation(s)
| | | | | | - Shuo Rainosek
- Department of Anesthesiology , Central Arkansas Veterans Health System , 4300 West Seventh Street, VA 704-110 , Little Rock , Arkansas 72205 , United States
| | | | | | | |
Collapse
|
43
|
Titz B, Gadaleta RM, Lo Sasso G, Elamin A, Ekroos K, Ivanov NV, Peitsch MC, Hoeng J. Proteomics and Lipidomics in Inflammatory Bowel Disease Research: From Mechanistic Insights to Biomarker Identification. Int J Mol Sci 2018; 19:ijms19092775. [PMID: 30223557 PMCID: PMC6163330 DOI: 10.3390/ijms19092775] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) represents a group of progressive disorders characterized by recurrent chronic inflammation of the gut. Ulcerative colitis and Crohn's disease are the major manifestations of IBD. While our understanding of IBD has progressed in recent years, its etiology is far from being fully understood, resulting in suboptimal treatment options. Complementing other biological endpoints, bioanalytical "omics" methods that quantify many biomolecules simultaneously have great potential in the dissection of the complex pathogenesis of IBD. In this review, we focus on the rapidly evolving proteomics and lipidomics technologies and their broad applicability to IBD studies; these range from investigations of immune-regulatory mechanisms and biomarker discovery to studies dissecting host⁻microbiome interactions and the role of intestinal epithelial cells. Future studies can leverage recent advances, including improved analytical methodologies, additional relevant sample types, and integrative multi-omics analyses. Proteomics and lipidomics could effectively accelerate the development of novel targeted treatments and the discovery of complementary biomarkers, enabling continuous monitoring of the treatment response of individual patients; this may allow further refinement of treatment and, ultimately, facilitate a personalized medicine approach to IBD.
Collapse
Affiliation(s)
- Bjoern Titz
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Raffaella M Gadaleta
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Giuseppe Lo Sasso
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Ashraf Elamin
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Irisviksvägen 31D, 02230 Esbo, Finland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, CH-2000 Neuchatel, Switzerland.
| |
Collapse
|
44
|
One- vs two-phase extraction: re-evaluation of sample preparation procedures for untargeted lipidomics in plasma samples. Anal Bioanal Chem 2018; 410:5859-5870. [PMID: 29968103 PMCID: PMC6096717 DOI: 10.1007/s00216-018-1200-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/24/2018] [Accepted: 06/14/2018] [Indexed: 12/02/2022]
Abstract
Lipidomics is a rapidly developing field in modern biomedical research. While LC-MS systems are able to detect most of the known lipid classes in a biological matrix, there is no single technique able to extract all of them simultaneously. In comparison with two-phase extractions, one-phase extraction systems are of particular interest, since they decrease the complexity of the experimental procedure. By using an untargeted lipidomics approach, we explored the differences/similarities between the most commonly used two-phase extraction systems (Folch, Bligh and Dyer, and MTBE) and one of the more recently introduced one-phase extraction systems for lipid analysis based on the MMC solvent mixture (MeOH/MTBE/CHCl3). The four extraction methods were evaluated and thoroughly compared against a pooled extract that qualitatively and quantitatively represents the average of the combined extractions. Our results show that the lipid profile obtained with the MMC system displayed the highest similarity to the pooled extract, indicating that it was most representative of the lipidome in the original sample. Furthermore, it showed better extraction efficiencies for moderate and highly apolar lipid species in comparison with the Folch, Bligh and Dyer, and MTBE extraction systems. Finally, the technical simplicity of the MMC procedure makes this solvent system highly suitable for automated, untargeted lipidomics analysis.
Collapse
|
45
|
Giles C, Takechi R, Lam V, Dhaliwal SS, Mamo JCL. Contemporary lipidomic analytics: opportunities and pitfalls. Prog Lipid Res 2018; 71:86-100. [PMID: 29959947 DOI: 10.1016/j.plipres.2018.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/18/2018] [Accepted: 06/26/2018] [Indexed: 01/08/2023]
Abstract
Recent advances in analytical techniques have greatly enhanced the depth of coverage, however lipidomic studies are still restricted to analysing only a subset of known lipids. Numerous complementary techniques are used for investigation of cellular lipidomes, including mass spectrometry (MS), nuclear magnetic resonance and vibrational spectroscopy. The development in electrospray ionization (ESI) MS has accelerated lipidomics research in the past two decades and represents one of the most widely used technique. The versatility of ESI-MS systems allows development of methods to detect and quantify a large diversity of lipid species and classes. However, highly targeted and specific approaches can preclude global analysis of many lipid classes. Indeed, experimental procedures are generally optimised for the lipid species, or lipid class of interest. Therefore, careful consideration of experimental procedures is required for characterisation of biological lipidomes. The current review will describe the lipidomic approaches for considering tissue lipid physiology. Discussion of the main sequences in a lipidomics workflow will be presented, including preparation of samples, accurate quantitation of lipid species and statistical modelling.
Collapse
Affiliation(s)
- Corey Giles
- Curtin Health Innovation Research Institute, Curtin University, WA, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, WA, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University, WA, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - Satvinder S Dhaliwal
- Curtin Health Innovation Research Institute, Curtin University, WA, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - John C L Mamo
- Curtin Health Innovation Research Institute, Curtin University, WA, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia.
| |
Collapse
|
46
|
Takeda H, Izumi Y, Takahashi M, Paxton T, Tamura S, Koike T, Yu Y, Kato N, Nagase K, Shiomi M, Bamba T. Widely-targeted quantitative lipidomics method by supercritical fluid chromatography triple quadrupole mass spectrometry. J Lipid Res 2018; 59:1283-1293. [PMID: 29724780 DOI: 10.1194/jlr.d083014] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/06/2018] [Indexed: 12/24/2022] Open
Abstract
Lipidomics, the mass spectrometry-based comprehensive analysis of lipids, has attracted attention as an analytical approach to provide novel insight into lipid metabolism and to search for biomarkers. However, an ideal method for both comprehensive and quantitative analysis of lipids has not been fully developed. Here, we have proposed a practical methodology for widely targeted quantitative lipidome analysis using supercritical fluid chromatography fast-scanning triple-quadrupole mass spectrometry (SFC/QqQMS) and theoretically calculated a comprehensive lipid multiple reaction monitoring (MRM) library. Lipid classes can be separated by SFC with a normal-phase diethylamine-bonded silica column with high resolution, high throughput, and good repeatability. Structural isomers of phospholipids can be monitored by mass spectrometric separation with fatty acyl-based MRM transitions. SFC/QqQMS analysis with an internal standard-dilution method offers quantitative information for both lipid class and individual lipid molecular species in the same lipid class. Additionally, data acquired using this method has advantages, including reduction of misidentification and acceleration of data analysis. Using the SFC/QqQMS system, alteration of plasma lipid levels in myocardial infarction-prone rabbits to the supplementation of EPA was first observed. Our developed SFC/QqQMS method represents a potentially useful tool for in-depth studies focused on complex lipid metabolism and biomarker discovery.-Takeda, H., Y. Izumi, M. Takahashi, T. Paxton, S. Tamura, T. Koike, Y. Yu, N. Kato, K. Nagase, M. Shiomi, and T. Bamba.
Collapse
Affiliation(s)
- Hiroaki Takeda
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Thanai Paxton
- Nihon Waters K.K., Shinagawa-ku, Tokyo 140-0001, Japan
| | - Shohei Tamura
- Institute of Experimental Animals, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Tomonari Koike
- Institute of Experimental Animals, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Ying Yu
- Institute of Experimental Animals, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Noriko Kato
- Nihon Waters K.K., Shinagawa-ku, Tokyo 140-0001, Japan
| | | | - Masashi Shiomi
- Institute of Experimental Animals, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
47
|
Sostare J, Di Guida R, Kirwan J, Chalal K, Palmer E, Dunn WB, Viant MR. Comparison of modified Matyash method to conventional solvent systems for polar metabolite and lipid extractions. Anal Chim Acta 2018; 1037:301-315. [PMID: 30292307 DOI: 10.1016/j.aca.2018.03.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/13/2018] [Accepted: 03/18/2018] [Indexed: 02/02/2023]
Abstract
In the last decade, metabolomics has experienced significant advances in the throughput and robustness of analytical methodologies. Yet the preparation of biofluids and low-mass tissue samples remains a laborious and potentially inconsistent manual process, and a significant bottleneck for high-throughput metabolomics. To address this, we have compared three different sample extraction solvent systems in three diverse sample types with the purpose of selecting an optimum protocol for subsequent automation of sample preparation. We have investigated and re-optimised the solvent ratios in the recently introduced methyl tert-butyl ether (MTBE)/methanol/water solvent system (here termed modified Matyash; 2.6/2.0/2.4, v/v/v) and compared it to the original Matyash method (10/3/2.5, v/v/v) and the conventional chloroform/methanol/water (stepwise Bligh and Dyer, 2.0/2.0/1.8, v/v/v) using two biofluids (human serum and urine) and one tissue (whole Daphnia magna). This is the first report of the use of the Matyash method for extracting metabolites from the US National Institutes of Health (NIH) model organism D. magna. Extracted samples were analysed by non-targeted direct infusion mass spectrometry metabolomics or LC-MS metabolomics. Overall, the modified Matyash method yielded a higher number of peaks and putatively annotated metabolites compared to the original Matyash method (1-29% more peaks and 1-30% more metabolites) and the Bligh and Dyer method (4-20% more peaks and 1-41% more metabolites). Additionally the modified Matyash method was superior when considering metabolite intensities. The reproducibility of the modified Matyash method was higher than other methods (in 10 out of 12 datasets, compared to the original Matyash method; and in 8 out of 12 datasets, compared to the Bligh and Dyer method), based upon the observation of a lower mRSD of peak intensities. In conclusion, the modified Matyash method tended to provide a higher yield and reproducibility for most sample types in this study compared to two widely used methods.
Collapse
Affiliation(s)
- Jelena Sostare
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Riccardo Di Guida
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Jennifer Kirwan
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Karnpreet Chalal
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Elliott Palmer
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Mark R Viant
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
48
|
Ali AS, Raju R, Ray S, Kshirsagar R, Gilbert A, Zang L, Karger BL. Lipidomics of CHO Cell Bioprocessing: Relation to Cell Growth and Specific Productivity of a Monoclonal Antibody. Biotechnol J 2018. [PMID: 29521466 DOI: 10.1002/biot.201700745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As the demand for biological therapeutic proteins rises, there is an increasing need for robust and highly efficient bioprocesses, specifically, maximizing protein production by controlling the cellular nutritional and metabolic needs. A comprehensive lipidomics analysis has been performed, for the first time, over the time course of CHO cells producing an IgG1 monoclonal antibody (mAb) with fed batch 5 L bioreactors. The dynamic nature and importance of the CHO lipidome, especially on cellular growth and specific productivity, is demonstrated. A robust LC-MS method using positive and negative mode ESI was developed for lipid identification and quantitation of 377 unique lipids. The analysis revealed large changes in lipid features between the different days in bioprocessing including accumulation of triacylglycerol (TG) and lysophospholipid species with depletion of diacylglycerol (DG) species. Exploring pathway analysis where the lipid data was combined with polar metabolites and transcriptomics (RNA sequencing) revealed differences in lipid metabolism between the various stages of cellular growth and highlighted the role of key features of lipid metabolism on cell growth and specific productivity. The study demonstrates the importance of lipidomics in the expanding role of 'Omics methodologies in gaining insight into cellular behavior during protein production in a fed batch bioprocess.
Collapse
Affiliation(s)
- Amr S Ali
- Cell Culture Development, Biogen, Inc., Cambridge, MA 02142, USA.,Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Ravali Raju
- Cell Culture Development, Biogen, Inc., Cambridge, MA 02142, USA
| | - Somak Ray
- Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | | | - Alan Gilbert
- Cell Culture Development, Biogen, Inc., Cambridge, MA 02142, USA
| | - Li Zang
- Analytical Development, Biogen, Inc., Cambridge, MA 02142, USA
| | - Barry L Karger
- Barnett Institute and Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
49
|
Abstract
The state-of-art in the lipidomic analysis is summarized here to provide the overview of available sample preparation strategies, mass spectrometry (MS)-based methods for the qualitative analysis of lipids, and the quantitative MS approaches for high-throughput clinical workflows. Major challenges in terms of widely accepted best practices for lipidomic analysis, nomenclature, and standards for data reporting are discussed as well.
Collapse
Affiliation(s)
- Michal Holčapek
- Department of Analytical Chemistry, Faculty of Chemical Technology , University of Pardubice , Studentská 573 , 53210 Pardubice , Czech Republic
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine , University Hospital Regensburg , 93053 Regensburg , Germany
| | - Kim Ekroos
- Lipidomics Consulting Ltd. , 02230 Esbo , Finland
| |
Collapse
|
50
|
Kavaliauskiene S, Torgersen ML, Lingelem ABD, Klokk TI, Lintonen T, Simolin H, Ekroos K, Skotland T, Sandvig K. Cellular effects of fluorodeoxyglucose: Global changes in the lipidome and alteration in intracellular transport. Oncotarget 2018; 7:79885-79900. [PMID: 27829218 PMCID: PMC5346758 DOI: 10.18632/oncotarget.13089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/21/2016] [Indexed: 11/29/2022] Open
Abstract
2-fluoro-2-deoxy-D-glucose (FDG), labeled with 18F radioisotope, is the most common imaging agent used for positron emission tomography (PET) in oncology. However, little is known about the cellular effects of FDG. Another glucose analogue, 2-deoxy-D-glucose (2DG), has been shown to affect many cellular functions, including intracellular transport and lipid metabolism, and has been found to improve the efficacy of cancer chemotherapeutic agents in vivo. Thus, in the present study, we have investigated cellular effects of FDG with the focus on changes in cellular lipids and intracellular transport. By quantifying more than 200 lipids from 17 different lipid classes in HEp-2 cells and by analyzing glycosphingolipids from MCF-7, HT-29 and HBMEC cells, we have discovered that FDG treatment inhibits glucosylceramide synthesis and thus reduces cellular levels of glycosphingolipids. In addition, in HEp-2 cells the levels and/or species composition of other lipid classes, namely diacylglycerols, phosphatidic acids and phosphatidylinositols, were found to change upon treatment with FDG. Furthermore, we show here that FDG inhibits retrograde Shiga toxin transport and is much more efficient in protecting cells against the toxin than 2DG. In summary, our data reveal novel effects of FDG on cellular transport and glycosphingolipid metabolism, which suggest a potential clinical application of FDG as an adjuvant for cancer chemotherapy.
Collapse
Affiliation(s)
- Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Anne Berit Dyve Lingelem
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Tove Irene Klokk
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | | | | | | | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|