1
|
Shannon CE, Bakewell T, Fourcaudot MJ, Ayala I, Smelter AA, Hinostroza EA, Romero G, Asmis M, Freitas Lima LC, Wallace M, Norton L. The mitochondrial pyruvate carrier regulates adipose glucose partitioning in female mice. Mol Metab 2024; 88:102005. [PMID: 39137831 PMCID: PMC11382204 DOI: 10.1016/j.molmet.2024.102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
OBJECTIVE The mitochondrial pyruvate carrier (MPC) occupies a critical node in intermediary metabolism, prompting interest in its utility as a therapeutic target for the treatment of obesity and cardiometabolic disease. Dysregulated nutrient metabolism in adipose tissue is a prominent feature of obesity pathophysiology, yet the functional role of adipose MPC has not been explored. We investigated whether the MPC shapes the adaptation of adipose tissue to dietary stress in female and male mice. METHODS The impact of pharmacological and genetic disruption of the MPC on mitochondrial pathways of triglyceride assembly (lipogenesis and glyceroneogenesis) was assessed in 3T3L1 adipocytes and murine adipose explants, combined with analyses of adipose MPC expression in metabolically compromised humans. Whole-body and adipose-specific glucose metabolism were subsequently investigated in male and female mice lacking adipocyte MPC1 (Mpc1AD-/-) and fed either standard chow, high-fat western style, or high-sucrose lipid restricted diets for 24 weeks, using a combination of radiolabeled tracers and GC/MS metabolomics. RESULTS Treatment with UK5099 or siMPC1 impaired the synthesis of lipids and glycerol-3-phosphate from pyruvate and blunted triglyceride accumulation in 3T3L1 adipocytes, whilst MPC expression in human adipose tissue was negatively correlated with indices of whole-body and adipose tissue metabolic dysfunction. Mature adipose explants from Mpc1AD-/- mice were intrinsically incapable of incorporating pyruvate into triglycerides. In vivo, MPC deletion restricted the incorporation of circulating glucose into adipose triglycerides, but only in female mice fed a zero fat diet, and this associated with sex-specific reductions in tricarboxylic acid cycle pool sizes and compensatory transcriptional changes in lipogenic and glycerol metabolism pathways. However, whole-body adiposity and metabolic health were preserved in Mpc1AD-/- mice regardless of sex, even under conditions of zero dietary fat. CONCLUSIONS These findings highlight the greater capacity for mitochondrially driven triglyceride assembly in adipose from female versus male mice and expose a reliance upon MPC-gated metabolism for glucose partitioning in female adipose under conditions of dietary lipid restriction.
Collapse
Affiliation(s)
- Christopher E Shannon
- UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland; Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
| | - Terry Bakewell
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Marcel J Fourcaudot
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Iriscilla Ayala
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Annie A Smelter
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Edgar A Hinostroza
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Giovanna Romero
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mara Asmis
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Leandro C Freitas Lima
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Martina Wallace
- UCD Conway Institute, School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Luke Norton
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
2
|
Duft RG, Bonfante ILP, Palma-Duran SA, Chacon-Mikahil MPT, Griffin JL, Cavaglieri CR. Moderate-intensity Combined Training Induces Lipidomic Changes in Individuals With Obesity and Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:2182-2198. [PMID: 38488044 PMCID: PMC11318996 DOI: 10.1210/clinem/dgae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 06/01/2024]
Abstract
CONTEXT Alterations in the lipid metabolism are linked to metabolic disorders such as insulin resistance (IR), obesity and type 2 diabetes (T2D). Regular exercise, particularly combined training (CT), is a well-known nonpharmacological treatment that combines aerobic (AT) and resistance (RT) training benefits. However, it is unclear whether moderate-intensity exercise without dietary intervention induces changes in lipid metabolism to promote a "healthy lipidome." OBJECTIVE The study aimed to investigate the effect of 16 weeks of CT on plasma and white adipose tissue in both sexes, middle-aged individuals with normal weight, obesity (OB), and T2D using an ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS) untargeted lipidomics approach. METHODS Body composition, maximum oxygen consumption (VO2max), strength, and biochemical markers were evaluated before and after the control/training period and correlated with lipid changes. CT consisted of 8 to 10 RT exercises, followed by 35 minutes of AT (45%-70% VO2max), 3 times a week for 16 weeks. RESULTS The CT significantly reduced the levels of saturated and monounsaturated fatty acid side-chains (SFA/MUFA) in sphingolipids, glycerolipids (GL) and glycerophospholipids (GP) as well as reducing fat mass, circumferences and IR. Increased levels of polyunsaturated fatty acids in GPs and GLs were also observed, along with increased fat-free mass, VO2 max, and strength (all P < .05) after training. CONCLUSION Our study revealed that 16 weeks of moderate-intensity CT remodeled the lipid metabolism in OB, and T2D individuals, even without dietary intervention, establishing a link between exercise-modulated lipid markers and mechanisms that reduce IR and obesity-related comorbidities.
Collapse
Affiliation(s)
- Renata Garbellini Duft
- Department of Metabolism, Digestion & Reproduction, Imperial College London, London SW7 2AZ, UK
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, 13083-851, São Paulo, Brazil
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Ivan Luiz Padilha Bonfante
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, 13083-851, São Paulo, Brazil
| | - Susana Alejandra Palma-Duran
- Department of Metabolism, Digestion & Reproduction, Imperial College London, London SW7 2AZ, UK
- Department of Food Science, Research Centre in Food and Development AC, Hermosillo, 83304, Mexico
| | | | - Julian Leether Griffin
- Department of Metabolism, Digestion & Reproduction, Imperial College London, London SW7 2AZ, UK
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Cláudia Regina Cavaglieri
- Laboratory of Exercise Physiology, Faculty of Physical Education, University of Campinas, 13083-851, São Paulo, Brazil
| |
Collapse
|
3
|
Shannon CE, Bakewell T, Fourcaudot MJ, Ayala I, Romero G, Asmis M, Lima LCF, Wallace M, Norton L. Sex-dependent adipose glucose partitioning by the mitochondrial pyruvate carrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593540. [PMID: 38798427 PMCID: PMC11118482 DOI: 10.1101/2024.05.11.593540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Objective The mitochondrial pyruvate carrier (MPC) occupies a critical node in intermediary metabolism, prompting interest in its utility as a therapeutic target for the treatment of obesity and cardiometabolic disease. Dysregulated nutrient metabolism in adipose tissue is a prominent feature of obesity pathophysiology, yet the functional role of adipose MPC has not been explored. We investigated whether the MPC shapes the adaptation of adipose tissue to dietary stress in female and male mice. Methods The impact of pharmacological and genetic disruption of the MPC on mitochondrial pathways of triglyceride assembly (lipogenesis and glyceroneogenesis) was assessed in 3T3L1 adipocytes and murine adipose explants, combined with analyses of adipose MPC expression in metabolically compromised humans. Whole-body and adipose-specific glucose metabolism were subsequently investigated in male and female mice lacking adipocyte MPC1 (Mpc1AD-/-) and fed either standard chow, high-fat western style, or high-sucrose lipid restricted diets for 24 weeks, using a combination of radiolabeled tracers and GC/MS metabolomics. Results Treatment with UK5099 or siMPC1 impaired the synthesis of lipids and glycerol-3-phosphate from pyruvate and blunted triglyceride accumulation in 3T3L1 adipocytes, whilst MPC expression in human adipose tissue was negatively correlated with indices of whole-body and adipose tissue metabolic dysfunction. Mature adipose explants from Mpc1AD-/- mice were intrinsically incapable of incorporating pyruvate into triglycerides. In vivo, MPC deletion restricted the incorporation of circulating glucose into adipose triglycerides, but only in female mice fed a zero fat diet, and this associated with sex-specific reductions in tricarboxylic acid cycle pool sizes and compensatory transcriptional changes in lipogenic and glycerol metabolism pathways. However, whole-body adiposity and metabolic health were preserved in Mpc1AD-/- mice regardless of sex, even under conditions of zero dietary fat. Conclusion These findings highlight the greater capacity for mitochondrially driven triglyceride assembly in adipose from female versus male mice and expose a reliance upon MPC-gated metabolism for glucose partitioning in female adipose under conditions of dietary lipid restriction.
Collapse
Affiliation(s)
- Christopher E Shannon
- UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Terry Bakewell
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Marcel J Fourcaudot
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Iriscilla Ayala
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Giovanna Romero
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mara Asmis
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Leandro C Freitas Lima
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Martina Wallace
- UCD Conway Institute, School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Luke Norton
- Diabetes Division, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
4
|
Qian C, Wang Q, Qiao Y, Xu Z, Zhang L, Xiao H, Lin Z, Wu M, Xia W, Yang H, Bai J, Geng D. Arachidonic acid in aging: New roles for old players. J Adv Res 2024:S2090-1232(24)00180-2. [PMID: 38710468 DOI: 10.1016/j.jare.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Arachidonic acid (AA), one of the most ubiquitous polyunsaturated fatty acids (PUFAs), provides fluidity to mammalian cell membranes. It is derived from linoleic acid (LA) and can be transformed into various bioactive metabolites, including prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs), hydroxy-eicosatetraenoic acids (HETEs), leukotrienes (LTs), and epoxyeicosatrienoic acids (EETs), by different pathways. All these processes are involved in AA metabolism. Currently, in the context of an increasingly visible aging world population, several scholars have revealed the essential role of AA metabolism in osteoporosis, chronic obstructive pulmonary disease, and many other aging diseases. AIM OF REVIEW Although there are some reviews describing the role of AA in some specific diseases, there seems to be no or little information on the role of AA metabolism in aging tissues or organs. This review scrutinizes and highlights the role of AA metabolism in aging and provides a new idea for strategies for treating aging-related diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW As a member of lipid metabolism, AA metabolism regulates the important lipids that interfere with the aging in several ways. We present a comprehensivereviewofthe role ofAA metabolism in aging, with the aim of relieving the extreme suffering of families and the heavy economic burden on society caused by age-related diseases. We also collected and summarized data on anti-aging therapies associated with AA metabolism, with the expectation of identifying a novel and efficient way to protect against aging.
Collapse
Affiliation(s)
- Chen Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Ze Xu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Linlin Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Zhixiang Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
5
|
Tang CH, Lin CY, Li HH, Kuo FW. Microplastics elicit an immune-agitative state in coral. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168406. [PMID: 37939952 DOI: 10.1016/j.scitotenv.2023.168406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/20/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
Microplastic pollution in the ocean is a major problem, as its pervasiveness elicits concerns the health impacts microplastics may have on marine life (such as reef-building corals). As a primary endpoint, the organismal lipidome can define the weakening of fitness and reveal the physiological context of adverse health effects in organisms. To gain insight into the effects of microplastics on coral health, lipid profiling was performed via an untargeted lipidomic approach on the coral Turbinaria mesenterina exposed to ~10 μm polystyrene microparticles for 10 days. Considerable microplastic accumulation and obvious effects relating with immune activation were observed in the coral treated with a near environmentally relevant concentration of microplastics (10 μg/L); however, these effects were not evident in the high level (100 μg/L) treatment group. In particular, increased levels of membrane lipids with 20:4 and 22:6 fatty acid chains reallocated from the triacylglycerol pool were observed in coral host cells and symbiotic algae, respectively, which could upregulate immune activity and realign symbiotic communication in coral. High levels of polyunsaturation can sensitize the coral cell membrane to lipid peroxidation and increase cell death, which is of greater concern; additionally, the photoprotective capacity of symbiotic algae was compromised. As a result, coral physiological functions were altered. These results show that, realistic levels of microplastic pollution can affect coral health and should be a concern.
Collapse
Affiliation(s)
- Chuan-Ho Tang
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan; Aerosol Science Research Center, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Ching-Yu Lin
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hsing-Hui Li
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Fu-Wen Kuo
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan
| |
Collapse
|
6
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
7
|
Palmgren H, Petkevicius K, Bartesaghi S, Ahnmark A, Ruiz M, Nilsson R, Löfgren L, Glover MS, Andréasson AC, Andersson L, Becquart C, Kurczy M, Kull B, Wallin S, Karlsson D, Hess S, Maresca M, Bohlooly-Y M, Peng XR, Pilon M. Elevated Adipocyte Membrane Phospholipid Saturation Does Not Compromise Insulin Signaling. Diabetes 2023; 72:1350-1363. [PMID: 36580483 PMCID: PMC10545576 DOI: 10.2337/db22-0293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022]
Abstract
Increased saturated fatty acid (SFA) levels in membrane phospholipids have been implicated in the development of metabolic disease. Here, we tested the hypothesis that increased SFA content in cell membranes negatively impacts adipocyte insulin signaling. Preadipocyte cell models with elevated SFA levels in phospholipids were generated by disrupting the ADIPOR2 locus, which resulted in a striking twofold increase in SFA-containing phosphatidylcholines and phosphatidylethanolamines, which persisted in differentiated adipocytes. Similar changes in phospholipid composition were observed in white adipose tissues isolated from the ADIPOR2-knockout mice. The SFA levels in phospholipids could be further increased by treating ADIPOR2-deficient cells with palmitic acid and resulted in reduced membrane fluidity and endoplasmic reticulum stress in mouse and human preadipocytes. Strikingly, increased SFA levels in differentiated adipocyte phospholipids had no effect on adipocyte gene expression or insulin signaling in vitro. Similarly, increased adipocyte phospholipid saturation did not impair white adipose tissue function in vivo, even in mice fed a high-saturated fat diet at thermoneutrality. We conclude that increasing SFA levels in adipocyte phospholipids is well tolerated and does not affect adipocyte insulin signaling in vitro and in vivo.
Collapse
Affiliation(s)
- Henrik Palmgren
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kasparas Petkevicius
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefano Bartesaghi
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrea Ahnmark
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mario Ruiz
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Ralf Nilsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Löfgren
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Matthew S. Glover
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Anne-Christine Andréasson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Liselotte Andersson
- Animal Science & Technologies, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Cécile Becquart
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Kurczy
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bengt Kull
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Simonetta Wallin
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Karlsson
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sonja Hess
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Marcello Maresca
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Ding L, Yang J, Guo H, Cong P, Xu J, Xue C, Mao X, Zhang T, Wang Y. Dietary Eicosapentaenoic Acid Containing Phosphoethanolamine Plasmalogens Remodels the Lipidome of White Adipose Tissue and Suppresses High-Fat Diet Induced Obesity in Mice. Mol Nutr Food Res 2023; 67:e2200321. [PMID: 37439463 DOI: 10.1002/mnfr.202200321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 04/26/2023] [Indexed: 07/14/2023]
Abstract
SCOPE Dietary supplementation of docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA) can alter the lipidome profiles of adipocytes, thereby counteract obesity. DHA/EPA in the form of phospholipids demonstrates higher bioavailability than triglyceride or ethyl ester (EE), but their effects on the lipidome and metabolic changes during obesity are still unknown. METHODS AND RESULTS High-fat diet-induced obese mice are treated with different molecular forms of EPA, and EPA supplemented as phosphoethanolamine plasmalogens (PlsEtn) has a superior effect on reducing fat mass accumulation than phosphatidylcholine (PC) or EE. The lipidomics analysis indicates that EPA in form of PlsEtn but not PC or EE significantly decreases total PC and sphingomyelin content in white adipose tissue (WAT). Some specific polyunsaturated fatty acid -containing PCs and ether phospholipids are increased in EPA-PlsEtn-fed mice, which may attribute to the upregulation of unsaturated fatty acid biosynthesis and fatty acid elongation reactions in WAT. In addition, the expression of genes related to fatty acid catabolism is also promoted by EPA-PlsEtn supplementation, which may cause the decreased content of saturated and monounsaturated fatty acid-containing PCs. CONCLUSIONS EPA-PlsEtn supplementation is demonstrated to remodel lipidome and regulate the fatty acid metabolic process in WAT, indicating it may serve as a new strategy for obesity treatment in the future.
Collapse
Affiliation(s)
- Lin Ding
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou, 215123, P. R. China
| | - Jinyue Yang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Haoran Guo
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou, 215123, P. R. China
| | - Peixu Cong
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Tiantian Zhang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, No.1299 Sansha Road, Qingdao, Shandong, 266000, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong, 266237, P. R. China
| |
Collapse
|
9
|
Cho YK, Lee S, Lee J, Doh J, Park JH, Jung YS, Lee YH. Lipid remodeling of adipose tissue in metabolic health and disease. Exp Mol Med 2023; 55:1955-1973. [PMID: 37653032 PMCID: PMC10545718 DOI: 10.1038/s12276-023-01071-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 09/02/2023] Open
Abstract
Adipose tissue is a dynamic and metabolically active organ that plays a crucial role in energy homeostasis and endocrine function. Recent advancements in lipidomics techniques have enabled the study of the complex lipid composition of adipose tissue and its role in metabolic disorders such as obesity, diabetes, and cardiovascular disease. In addition, adipose tissue lipidomics has emerged as a powerful tool for understanding the molecular mechanisms underlying these disorders and identifying bioactive lipid mediators and potential therapeutic targets. This review aims to summarize recent lipidomics studies that investigated the dynamic remodeling of adipose tissue lipids in response to specific physiological changes, pharmacological interventions, and pathological conditions. We discuss the molecular mechanisms of lipid remodeling in adipose tissue and explore the recent identification of bioactive lipid mediators generated in adipose tissue that regulate adipocytes and systemic metabolism. We propose that manipulating lipid-mediator metabolism could serve as a therapeutic approach for preventing or treating obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yoon Keun Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sumin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaewon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX Institute, Soft Foundry Institute, Seoul National University, Seoul, Republic of Korea
| | - Joo-Hong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Soria-Gondek A, Fernández-García P, González L, Reyes-Farias M, Murillo M, Valls A, Real N, Pellitero S, Tarascó J, Jenkins B, Galán M, Villarroya F, Koulman A, Corrales P, Vidal-Puig A, Cereijo R, Sánchez-Infantes D. Lipidome Profiling in Childhood Obesity Compared to Adults: A Pilot Study. Nutrients 2023; 15:3341. [PMID: 37571279 PMCID: PMC10421258 DOI: 10.3390/nu15153341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/13/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The objective is to assess the circulating lipidome of children with obesity before and after lifestyle intervention and to compare the data to the circulating lipidome of adults with obesity before and after bariatric surgery. Ten pediatric (PE) and thirty adult (AD) patients with obesity were prospectively recruited at a referral single center. The PE cohort received lifestyle recommendations. The AD cohort underwent bariatric surgery. Clinical parameters and lipidome were analyzed in serum before and after six months of metabolic intervention. The abundance of phosphatidylinositols in the PE cohort and phosphatidylcholines in the AD significantly increased, while O-phosphatidylserines in the PE cohort and diacyl/triacylglycerols in the AD decreased. Fifteen lipid species were coincident in both groups after lifestyle intervention and bariatric surgery. Five species of phosphatidylinositols, sphingomyelins, and cholesteryl esters were upregulated. Eight species of diacylglycerols, glycerophosphoglycerols, glycerophosphoethanolamines, and phosphatidylcholines were downregulated. Most matching species were regulated in the same direction except for two phosphatidylinositols: PI(O-36:2) and PI(O-34:0). A specific set of lipid species regulated after bariatric surgery in adult individuals was also modulated in children undergoing lifestyle intervention, suggesting they may constitute a core circulating lipid profile signature indicative of early development of obesity and improvement after clinical interventions regardless of individual age.
Collapse
Affiliation(s)
- Andrea Soria-Gondek
- Pediatric Surgery Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Pablo Fernández-García
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Lorena González
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
| | - Marjorie Reyes-Farias
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Marta Murillo
- Pediatric Endocrinology Unit, Pediatric Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (M.M.); (A.V.)
| | - Aina Valls
- Pediatric Endocrinology Unit, Pediatric Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (M.M.); (A.V.)
| | - Nativitat Real
- Pediatric Nurse, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Silvia Pellitero
- Endocrinology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Jordi Tarascó
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1GG, UK; (B.J.); (A.K.)
| | - María Galán
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Francesc Villarroya
- Biochemistry and Molecular Biomedicine Department, Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1GG, UK; (B.J.); (A.K.)
| | - Patricia Corrales
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 1GG, UK;
| | - Rubén Cereijo
- Biochemistry and Molecular Biomedicine Department, Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - David Sánchez-Infantes
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
| |
Collapse
|
11
|
Šarac I, Debeljak-Martačić J, Takić M, Stevanović V, Milešević J, Zeković M, Popović T, Jovanović J, Vidović NK. Associations of fatty acids composition and estimated desaturase activities in erythrocyte phospholipids with biochemical and clinical indicators of cardiometabolic risk in non-diabetic Serbian women: the role of level of adiposity. Front Nutr 2023; 10:1065578. [PMID: 37545582 PMCID: PMC10397414 DOI: 10.3389/fnut.2023.1065578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Fatty acids (FAs) composition and desaturase activities can be altered in different metabolic conditions, but the adiposity-independent associations with clinical and biochemical indicators of cardiometabolic risk are still unclear. This study aimed to analyze the associations of FAs composition and estimated desaturase activities with anthropometric, clinical, and biochemical cardiometabolic risk indicators in non-diabetic Serbian women, and to investigate if these associations were independent of the level of adiposity and other confounders. Methods In 76 non-diabetic, otherwise healthy Serbian women, aged 24-68 years, with or without metabolic syndrome or obesity (BMI=23.6±5.6 kg/m2), FA composition in erythrocyte phospholipids was measured by gas-liquid chromatography. Desaturase activities were estimated from product/precursor FAs ratios (D9D:16:1n-7/16:0; D6D:20:3n-6/18:2n-6; D5D:20:4n-6/20:3n-6). Correlations were made with anthropometric, biochemical (serum glucose, triacylglycerols, LDL-C, HDL-C, ALT, AST, and their ratios) and clinical (blood pressure) indicators of cardiometabolic risk. Linear regression models were performed to test the independence of these associations. Results Estimated desaturase activities and certain FAs were associated with anthropometric, clinical and biochemical indicators of cardiometabolic risk: D9D, D6D, 16:1n-7 and 20:3n-6 were directly associated, while D5D and 18:0 were inversely associated. However, the associations with clinical and biochemical indicators were not independent of the associations with the level of adiposity, since they were lost after controlling for anthropometric indices. After controlling for multiple confounders (age, postmenopausal status, education, smoking, physical activity, dietary macronutrient intakes, use of supplements, alcohol consumption), the level of adiposity was the most significant predictor of desaturase activities and aforementioned FAs levels, and mediated their association with biochemical/clinical indicators. Vice versa, desaturase activities predicted the level of adiposity, but not other components of cardiometabolic risk (if the level of adiposity was accounted). While the associations of anthropometric indices with 16:1n-7, 20:3n-6, 18:0 and D9D and D6D activities were linear, the associations with D5D activity were the inverse U-shaped. The only adiposity-independent association of FAs profiles with the indicators of cardiometabolic risk was a positive association of 20:5n-3 with ALT/AST ratio, which requires further exploration. Discussion Additional studies are needed to explore the mechanisms of the observed associations.
Collapse
Affiliation(s)
- Ivana Šarac
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Debeljak-Martačić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Takić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vuk Stevanović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Milešević
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Popović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jovica Jovanović
- Department of Occupational Health, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Nevena Kardum Vidović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
12
|
Zheng R, Michaëlsson K, Fall T, Elmståhl S, Lind L. The metabolomic profiling of total fat and fat distribution in a multi-cohort study of women and men. Sci Rep 2023; 13:11129. [PMID: 37429905 DOI: 10.1038/s41598-023-38318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Currently studies aiming for the comprehensive metabolomics profiling of measured total fat (%) as well as fat distribution in both sexes are lacking. In this work, bioimpedance analysis was applied to measure total fat (%) and fat distribution (trunk to leg ratio). Liquid chromatography-mass spectrometry-based untargeted metabolomics was employed to profile the metabolic signatures of total fat (%) and fat distribution in 3447 participants from three Swedish cohorts (EpiHealth, POEM and PIVUS) using a discovery-replication cross-sectional study design. Total fat (%) and fat distribution were associated with 387 and 120 metabolites in the replication cohort, respectively. Enriched metabolic pathways for both total fat (%) and fat distribution included protein synthesis, branched-chain amino acids biosynthesis and metabolism, glycerophospholipid metabolism and sphingolipid metabolism. Four metabolites were mainly related to fat distribution: glutarylcarnitine (C5-DC), 6-bromotryptophan, 1-stearoyl-2-oleoyl-GPI (18:0/18:1) and pseudouridine. Five metabolites showed different associations with fat distribution in men and women: quinolinate, (12Z)-9,10-dihydroxyoctadec-12-enoate (9,10-DiHOME), two sphingomyelins and metabolonic lactone sulfate. To conclude, total fat (%) and fat distribution were associated with a large number of metabolites, but only a few were exclusively associated with fat distribution and of those metabolites some were associated with sex*fat distribution. Whether these metabolites mediate the undesirable effects of obesity on health outcomes remains to be further investigated.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - Karl Michaëlsson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sölve Elmståhl
- Division of Geriatric Medicine, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Kimura T, Kimura AK, Epand RM. Systematic crosstalk in plasmalogen and diacyl lipid biosynthesis for their differential yet concerted molecular functions in the cell. Prog Lipid Res 2023; 91:101234. [PMID: 37169310 DOI: 10.1016/j.plipres.2023.101234] [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: 03/03/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
Plasmalogen is a major phospholipid of mammalian cell membranes. Recently it is becoming evident that the sn-1 vinyl-ether linkage in plasmalogen, contrasting to the ester linkage in the counterpart diacyl glycerophospholipid, yields differential molecular characteristics for these lipids especially related to hydrocarbon-chain order, so as to concertedly regulate biological membrane processes. A role played by NMR in gaining information in this respect, ranging from molecular to tissue levels, draws particular attention. We note here that a broad range of enzymes in de novo synthesis pathway of plasmalogen commonly constitute that of diacyl glycerophospholipid. This fact forms the basis for systematic crosstalk that not only controls a quantitative balance between these lipids, but also senses a defect causing loss of lipid in either pathway for compensation by increase of the counterpart lipid. However, this inherent counterbalancing mechanism paradoxically amplifies imbalance in differential effects of these lipids in a diseased state on membrane processes. While sharing of enzymes has been recognized, it is now possible to overview the crosstalk with growing information for specific enzymes involved. The overview provides a fundamental clue to consider cell and tissue type-dependent schemes in regulating membrane processes by plasmalogen and diacyl glycerophospholipid in health and disease.
Collapse
Affiliation(s)
- Tomohiro Kimura
- Department of Chemistry & Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, USA.
| | - Atsuko K Kimura
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
14
|
Mocciaro G, Allison M, Jenkins B, Azzu V, Huang-Doran I, Herrera-Marcos LV, Hall Z, Murgia A, Susan D, Frontini M, Vidal-Puig A, Koulman A, Griffin JL, Vacca M. Non-alcoholic fatty liver disease is characterised by a reduced polyunsaturated fatty acid transport via free fatty acids and high-density lipoproteins (HDL). Mol Metab 2023; 73:101728. [PMID: 37084865 PMCID: PMC10176260 DOI: 10.1016/j.molmet.2023.101728] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/25/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) develops due to impaired hepatic lipid fluxes and is a risk factor for chronic liver disease and atherosclerosis. Lipidomic studies consistently reported characteristic hepatic/VLDL "lipid signatures" in NAFLD; whole plasma traits are more debated. Surprisingly, the HDL lipid composition by mass spectrometry has not been characterised across the NAFLD spectrum, despite HDL being a possible source of hepatic lipids delivered from peripheral tissues alongside free fatty acids (FFA). This study characterises the HDL lipidomic signature in NAFLD, and its correlation with metabolic and liver disease markers. METHODS We used liquid chromatography-mass spectrometry to determine the whole serum and HDL lipidomic profile in 89 biopsy-proven NAFLD patients and 20 sex and age-matched controls. RESULTS In the whole serum of NAFLD versus controls, we report a depletion in polyunsaturated (PUFA) phospholipids (PL) and FFA; with PUFA PL being also lower in HDL, and negatively correlated with BMI, insulin resistance, triglycerides, and hepatocyte ballooning. In the HDL of the NAFLD group we also describe higher saturated ceramides, which positively correlate with insulin resistance and transaminases. CONCLUSION NAFLD features lower serum lipid species containing polyunsaturated fatty acids; the most affected lipid fractions are FFA and (HDL) phospholipids; our data suggest a possible defect in the transfer of PUFA from peripheral tissues to the liver in NAFLD. Mechanistic studies are required to explore the biological implications of our findings addressing if HDL composition can influence liver metabolism and damage, thus contributing to NAFLD pathophysiology.
Collapse
Affiliation(s)
- Gabriele Mocciaro
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom
| | - Michael Allison
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Benjamin Jenkins
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Vian Azzu
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Isabel Huang-Doran
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Luis Vicente Herrera-Marcos
- Department of Biochemistry and Molecular and Cellular Biology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| | - Zoe Hall
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Antonio Murgia
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom
| | - Davies Susan
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Mattia Frontini
- Faculty of Health and Life Sciences, Clinical and Biomedical Sciences, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - Antonio Vidal-Puig
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Albert Koulman
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom.
| | - Julian L Griffin
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; The Rowett Institute, Foresterhill Campus, University of Aberdeen, Aberdeen, AB25 2ZD, United Kingdom.
| | - Michele Vacca
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom; Aldo Moro University of Bari, Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", Bari, 70124, Italy.
| |
Collapse
|
15
|
Li X, Guo Y, Wang X, Li H, Mao J, Yan S, Luo G, Wang R, Wu X, Li Y. Seminal plasma metabolomics signatures of normosmic congenital hypogonadotropic hypogonadism. Heliyon 2023; 9:e14779. [PMID: 37025907 PMCID: PMC10070097 DOI: 10.1016/j.heliyon.2023.e14779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 03/02/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Background Normosmic congenital hypogonadotropic hypogonadism (nCHH) is a rare disease, whose pathogenesis remains unclear. Here, we conducted untargeted metabolomics and lipidomics to identify seminal plasma signatures of nCHH, and to study the effect of LH and FSH deficiency on semen. Methods Twenty-five diagnosed patients with nCHH (HH group) and twenty-three healthy participants (HC group) were enrolled. Laboratory parameters, seminal plasma samples and patients' medical data were collected. Untargeted metabolomics and lipidomic profiling were performed using mass spectrometry (MS). Results The metabolomics profiling are altered among patients with nCHH and healthy controls. There are 160 kinds of differential metabolites and the main different lipid species are TAG, PC, SM and PE. Conclusions The metabolomics profiles in patients with nCHH changed. We hope that this work provides important insights into the pathophysiology of nCHH.
Collapse
Affiliation(s)
- Xiaogang Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ye Guo
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xi Wang
- National Health Commission Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jiangfeng Mao
- National Health Commission Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Songxin Yan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoju Luo
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Corresponding author. Department of Neurosurgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Xueyan Wu
- National Health Commission Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Corresponding authors. Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuai Fuyuan, Dong Cheng District, Beijing, 100730, China.
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Corresponding author. Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China.
| |
Collapse
|
16
|
Blandin A, Dugail I, Hilairet G, Ponnaiah M, Ghesquière V, Froger J, Ducheix S, Fizanne L, Boursier J, Cariou B, Lhomme M, Le Lay S. Lipidomic analysis of adipose-derived extracellular vesicles reveals specific EV lipid sorting informative of the obesity metabolic state. Cell Rep 2023; 42:112169. [PMID: 36862553 DOI: 10.1016/j.celrep.2023.112169] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
Adipose extracellular vesicles (AdEVs) transport lipids that could participate in the development of obesity-related metabolic dysfunctions. This study aims to define mouse AdEV lipid signature by a targeted LC-MS/MS approach in either healthy or obesity context. Distinct clustering of AdEV and visceral adipose tissue (VAT) lipidomes by principal component analysis reveals specific AdEV lipid sorting when compared with secreting VAT. Comprehensive analysis identifies enrichment of ceramides, sphingomyelins, and phosphatidylglycerols species in AdEVs compared with source VAT whose lipid content closely relates to the obesity status and is influenced by the diet. Obesity moreover impacts AdEV lipidome, mirroring lipid alterations retrieved in plasma and VAT. Overall, our study identifies specific lipid fingerprints for plasma, VAT, and AdEVs that are informative of the metabolic status. Lipid species enriched in AdEVs in the obesity context may constitute biomarker candidates or mediators of the obesity-associated metabolic dysfunctions.
Collapse
Affiliation(s)
- Alexia Blandin
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Isabelle Dugail
- UMRS 1269 INSERM/Sorbonne University, Nutriomics, 75013 Paris, France
| | | | - Maharajah Ponnaiah
- IHU ICAN (ICAN Omics and ICAN I/O), Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Valentine Ghesquière
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Josy Froger
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Simon Ducheix
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Lionel Fizanne
- HIFIH Laboratory UPRES EA3859, SFR 4208, Angers University, Angers, France
| | - Jérôme Boursier
- HIFIH Laboratory UPRES EA3859, SFR 4208, Angers University, Angers, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Marie Lhomme
- IHU ICAN (ICAN Omics and ICAN I/O), Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Soazig Le Lay
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France.
| |
Collapse
|
17
|
Gertner DS, Violi JP, Bishop DP, Padula MP. Lipid Spectrum Generator: A Simple Script for the Generation of Accurate In Silico Lipid Fragmentation Spectra. Anal Chem 2023; 95:2909-2916. [PMID: 36692449 DOI: 10.1021/acs.analchem.2c04518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Due to the complexity of lipids in nature, the use of in silico generated spectral libraries to identify lipid species from mass spectral data has become an integral part of many lipidomic workflows. However, many in silico libraries are either limited in usability or their capacity to represent lipid species. Here, we introduce Lipid Spectrum Generator, an open-source in silico spectral library generator specifically designed to aid in the identification of lipids in liquid chromatography-tandem mass spectrometry analysis.
Collapse
Affiliation(s)
- David S Gertner
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Ultimo 2007, Australia
| | - Jake P Violi
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Ultimo 2007, Australia
| | - David P Bishop
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo 2007, Australia
| | - Matthew P Padula
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Ultimo 2007, Australia
| |
Collapse
|
18
|
Kong Y, Jiang J, Huang Y, Liu X, Jin Z, Li L, Wei F, Liu X, Yin J, Zhang Y, Tong Q, Chen H. Narciclasine inhibits phospholipase A2 and regulates phospholipid metabolism to ameliorate psoriasis-like dermatitis. Front Immunol 2023; 13:1094375. [PMID: 36700214 PMCID: PMC9869703 DOI: 10.3389/fimmu.2022.1094375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Psoriasis is a common inflammatory skin disease recognized by the World Health Organization as "an incurable chronic, noninfectious, painful, disfiguring and disabling disease." The fact that metabolic syndrome (MetS) is the most common and important comorbidities of psoriasis suggests an important role of lipid metabolism in the pathogenesis of psoriasis. Narciclasine (Ncs) is an alkaloid isolated from the Amaryllidaceae plants. Its biological activities include antitumor, antibacterial, antiinflammatory, anti-angiogenic and promoting energy expenditure to improve dietinduced obesity. Here, we report that Ncs may be a potential candidate for psoriasis, acting at both the organismal and cellular levels. Methods The therapeutic effect of Ncs was assessed in IMQ-induced psoriasis-like mouse model. Then, through in vitro experiments, we explored the inhibitory effect of Ncs on HaCaT cell proliferation and Th17 cell polarization; Transcriptomics and lipidomics were used to analyze the major targets of Ncs; Single-cell sequencing data was used to identify the target cells of Ncs action. Results Ncs can inhibit keratinocyte proliferation and reduce the recruitment of immune cells in the skin by inhibiting psoriasis-associated inflammatory mediators. In addition, it showed a direct repression effect on Th17 cell polarization. Transcriptomic and lipidomic data further revealed that Ncs extensively regulated lipid metabolismrelated genes, especially the Phospholipase A2 (PLA2) family, and increased antiinflammatory lipid molecules. Combined with single-cell data analysis, we confirmed that keratinocytes are the main cells in which Ncs functions. Discussion Taken together, our findings indicate that Ncs alleviates psoriasiform skin inflammation in mice, which is associated with inhibition of PLA2 in keratinocytes and improved phospholipid metabolism. Ncs has the potential for further development as a novel anti-psoriasis drug.
Collapse
Affiliation(s)
- Yi Kong
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqiong Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zilin Jin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Li
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi, China
| | - Fen Wei
- Department of Dermatology, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, Guangdong, China
| | - Xinxin Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Yin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji-Rongcheng Center for Biomedicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji-Rongcheng Center for Biomedicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Hongxiang Chen, ; Qingyi Tong, ; Yonghui Zhang,
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji-Rongcheng Center for Biomedicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Hongxiang Chen, ; Qingyi Tong, ; Yonghui Zhang,
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,Department of Dermatology, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, Guangdong, China,*Correspondence: Hongxiang Chen, ; Qingyi Tong, ; Yonghui Zhang,
| |
Collapse
|
19
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Supplementing Diets with Agriophyllum squarrosum Reduced Blood Lipids, Enhanced Immunity and Anti-Inflammatory Capacities, and Mediated Lipid Metabolism in Tan Lambs. Animals (Basel) 2022; 12:ani12243486. [PMID: 36552407 PMCID: PMC9774518 DOI: 10.3390/ani12243486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022] Open
Abstract
Agriophyllum squarrosum (sand rice), a widespread desert plant, possesses anti-hyperglycemic and anti-inflammatory properties, and has been used in traditional Chinese medicine for many years. However, its effects on ruminants are unknown. To fill this gap, we examined the effects of A. squarrosum on the immune and anti-inflammatory responses of lambs. A total of 23, 6-month-old Tan ewe-lambs (27.6 ± 0.47 kg) were divided into four groups and offered a basic diet (C—control), or a diet that contained 10%, 20%, or 30% A. squarrosum, on a dry matter basis, for 128 days. Serum concentrations of total cholesterol were lower (p = 0.004) in the 30% supplemented lambs than controls, while concentrations of high-density lipoprotein cholesterol were lower (p = 0.006) in the 10% and 20%, but not in 30% supplemented lambs than controls. Serum-cortisol concentrations were lower (p = 0.012) in the 30% supplemented lambs and free fatty acid concentrations were higher in the 10% and 20% supplemented lambs than in control lambs (p < 0.001). Supplementation with A. squarrosum decreased (p < 0.05) the area of adipocytes in subcutaneous adipose tissue, but there was no difference between the 20% and 30% diets. Conversely, the area in visceral adipose tissue (VAT) increased (p < 0.05), especially for the 10% and 20% supplemented diets. Supplementation with A. squarrosum also enriched immune and anti-inflammatory related and lipid and glucose-metabolic pathways and associated differentially expressed gene expressions in adipose tissue. A total of 10 differential triacylglycerol, 34 differential phosphatidylcholines and seven differential phosphatidylethanolamines decreased in the diet with 30% supplementation, when compared to the other diets. Finally, adipocyte-differentiation genes, and immune and inflammatory response-related gene expression levels decreased in lamb adipocytes cultured with an aqueous A. squarrosum extract. In conclusion, supplementing lamb diets with A. squarrosum reduced blood lipids, enhanced immunity and anti-inflammatory capacities, and mediated lipid metabolism in adipose tissue and adipocytes of Tan lambs. A level of approximately 10% is recommended, but further research is required to determine the precise optimal level.
Collapse
|
21
|
Kleiboeker B, Lodhi IJ. Peroxisomal regulation of energy homeostasis: Effect on obesity and related metabolic disorders. Mol Metab 2022; 65:101577. [PMID: 35988716 PMCID: PMC9442330 DOI: 10.1016/j.molmet.2022.101577] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Peroxisomes are single membrane-bound organelles named for their role in hydrogen peroxide production and catabolism. However, their cellular functions extend well beyond reactive oxygen species (ROS) metabolism and include fatty acid oxidation of unique substrates that cannot be catabolized in mitochondria, and synthesis of ether lipids and bile acids. Metabolic functions of peroxisomes involve crosstalk with other organelles, including mitochondria, endoplasmic reticulum, lipid droplets and lysosomes. Emerging studies suggest that peroxisomes are important regulators of energy homeostasis and that disruption of peroxisomal functions influences the risk for obesity and the associated metabolic disorders, including type 2 diabetes and hepatic steatosis. SCOPE OF REVIEW Here, we focus on the role of peroxisomes in ether lipid synthesis, β-oxidation and ROS metabolism, given that these functions have been most widely studied and have physiologically relevant implications in systemic metabolism and obesity. Efforts are made to mechanistically link these cellular and systemic processes. MAJOR CONCLUSIONS Circulating plasmalogens, a form of ether lipids, have been identified as inversely correlated biomarkers of obesity. Ether lipids influence metabolic homeostasis through multiple mechanisms, including regulation of mitochondrial morphology and respiration affecting brown fat-mediated thermogenesis, and through regulation of adipose tissue development. Peroxisomal β-oxidation also affects metabolic homeostasis through generation of signaling molecules, such as acetyl-CoA and ROS that inhibit hydrolysis of stored lipids, contributing to development of hepatic steatosis. Oxidative stress resulting from increased peroxisomal β-oxidation-generated ROS in the context of obesity mediates β-cell lipotoxicity. A better understanding of the roles peroxisomes play in regulating and responding to obesity and its complications will provide new opportunities for their treatment.
Collapse
Affiliation(s)
- Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA.
| |
Collapse
|
22
|
Simond AÉ, Noël M, Loseto L, Houde M, Kirk J, Elliott A, Brown TM. A Multi-Matrix Metabolomic Approach in Ringed Seals and Beluga Whales to Evaluate Contaminant and Climate-Related Stressors. Metabolites 2022; 12:813. [PMID: 36144217 PMCID: PMC9502077 DOI: 10.3390/metabo12090813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
As a high trophic-level species, ringed seals (Pusa hispida) and beluga whales (Delphinapterus leucas) are particularly vulnerable to elevated concentrations of biomagnifying contaminants, such as polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs) and mercury (Hg). These species also face climate-change-related impacts which are leading to alterations in their diet and associated contaminant exposure. The metabolomic profile of marine mammal tissues and how it changes to environmental stressors is poorly understood. This study characterizes the profiles of 235 metabolites across plasma, liver, and inner and outer blubber in adult ringed seals and beluga whales and assesses how these profiles change as a consequence of contaminants and dietary changes. In both species, inner and outer blubber were characterized by a greater proportion of lipid classes, whereas the dominant metabolites in liver and plasma were amino acids, carbohydrates, biogenic amines and lysophosphatidylcholines. Several metabolite profiles in ringed seal plasma correlated with δ13C, while metabolite profiles in blubber were affected by hexabromobenzene in ringed seals and PBDEs and Hg in belugas. This study provides insight into inter-matrix similarities and differences across tissues and suggests that plasma and liver are more suitable for studying changes in diet, whereas liver and blubber are more suitable for studying the impacts of contaminants.
Collapse
Affiliation(s)
- Antoine É. Simond
- Pacific Science Enterprise Centre, Fisheries and Oceans Canada, 4160 Marine Drive, West Vancouver, BC V7V 1N6, Canada
- School of Resource and Environmental Management, Simon Fraser University, 4160 Marine Drive, West Vancouver, BC V7V 1N6, Canada
| | - Marie Noël
- Ocean Wise, 101-440 Cambie Street, Vancouver, BC V6B 2N5, Canada
| | - Lisa Loseto
- Freshwater Institute, Fisheries and Oceans Canada, 501 University Crescent, Winnipeg, MB R3T 2N6, Canada
- Centre for Earth Observation Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Magali Houde
- Centre St-Laurent, Environment and Climate Change Canada, 105 McGill Street, Montreal, QC H2Y 2E7, Canada
| | - Jane Kirk
- Canada Centre for Inland Waters, Environment and Climate Change Canada, 867 Lakeshore Road, Burlington, ON L7S 1A1, Canada
| | - Ashley Elliott
- Freshwater Institute, Fisheries and Oceans Canada, 501 University Crescent, Winnipeg, MB R3T 2N6, Canada
| | - Tanya M. Brown
- Pacific Science Enterprise Centre, Fisheries and Oceans Canada, 4160 Marine Drive, West Vancouver, BC V7V 1N6, Canada
- School of Resource and Environmental Management, Simon Fraser University, 4160 Marine Drive, West Vancouver, BC V7V 1N6, Canada
| |
Collapse
|
23
|
Meneses MJ, Sousa-Lima I, Jarak I, Raposo JF, Alves MG, Macedo MP. Distinct impacts of fat and fructose on the liver, muscle, and adipose tissue metabolome: An integrated view. Front Endocrinol (Lausanne) 2022; 13:898471. [PMID: 36060961 PMCID: PMC9428722 DOI: 10.3389/fendo.2022.898471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Objective In the last years, changes in dietary habits have contributed to the increasing prevalence of metabolic disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). The differential burden of lipids and fructose on distinct organs needs to be unveiled. Herein, we hypothesized that high-fat and high-fructose diets differentially affect the metabolome of insulin-sensitive organs such as the liver, muscle, and different adipose tissue depots. Methods We have studied the impact of 12 weeks of a control (11.50% calories from fat, 26.93% from protein, and 61.57% from carbohydrates), high-fat/sucrose (HFat), or high-fructose (HFruct) feeding on C57Bl/6J male mice. Besides glucose homeostasis, we analyzed the hepatic levels of glucose and lipid-metabolism-related genes and the metabolome of the liver, the muscle, and white (WAT) and brown adipose tissue (BAT) depots. Results HFat diet led to a more profound impact on hepatic glucose and lipid metabolism than HFruct, with mice presenting glucose intolerance, increased saturated fatty acids, and no glycogen pool, yet both HFat and HFruct presented hepatic insulin resistance. HFat diet promoted a decrease in glucose and lactate pools in the muscle and an increase in glutamate levels. While HFat had alterations in BAT metabolites that indicate increased thermogenesis, HFruct led to an increase in betaine, a protective metabolite against fructose-induced inflammation. Conclusions Our data illustrate that HFat and HFruct have a negative but distinct impact on the metabolome of the liver, muscle, WAT, and BAT.
Collapse
Affiliation(s)
- Maria João Meneses
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Inês Sousa-Lima
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - João F. Raposo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Marco G. Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
- Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
24
|
Guo Y, Li X, Wang X, Li H, Luo G, Si Y, Wu X, Li Y. Seminal plasma metabolomics and lipidomics profiling to identify signatures of pituitary stalk interruption syndrome. Orphanet J Rare Dis 2022; 17:267. [PMID: 35841106 PMCID: PMC9287950 DOI: 10.1186/s13023-022-02408-4] [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: 01/13/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background Pituitary stalk interruption syndrome (PSIS) is a rare disease caused by congenital pituitary anatomical defects. The underlying mechanisms remain unclear, and the diagnosis is difficult. Here, integrated metabolomics and lipidomics profiling were conducted to study the pathogenesis of PSIS. Methods Twenty-one patients with PSIS (BD group) and twenty-three healthy controls (HC group) were enrolled. Basal information and seminal plasma samples were collected. Untargeted metabolomics and lipidomics analyses were performed using ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS). Results The metabolomics and lipidomics profiles of patients with PSIS changed. The prolactin signaling pathway and biosynthesis of amino acids were the main differentially modified metabolic pathways. The main differentially modified metabolites were triacylglycerols (TGs), phosphatidylethanolamine (PE), sphingomyelin (SM), ceramide (Cer) and phosphatidylcholines (PCs). Pregnenolones and L-saccharopine could achieve a diagnosis of PSIS. Conclusions Pregnenolones and L-saccharopine are potential biomarkers for a PSIS diagnosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02408-4.
Collapse
Affiliation(s)
- Ye Guo
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China
| | - Xiaogang Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China.,Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xi Wang
- National Health Commission Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuai Fuyuan, Dong Cheng District, Beijing, 100730, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China
| | - Guoju Luo
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China
| | - Yongzhen Si
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China
| | - Xueyan Wu
- National Health Commission Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuai Fuyuan, Dong Cheng District, Beijing, 100730, China.
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 1 Shuaifuyuan Road, Beijing, 100730, China.
| |
Collapse
|
25
|
Adipocyte lysoplasmalogenase TMEM86A regulates plasmalogen homeostasis and protein kinase A-dependent energy metabolism. Nat Commun 2022; 13:4084. [PMID: 35835749 PMCID: PMC9283435 DOI: 10.1038/s41467-022-31805-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of adipose tissue plasmalogen metabolism is associated with obesity-related metabolic diseases. We report that feeding mice a high-fat diet reduces adipose tissue lysoplasmalogen levels and increases transmembrane protein 86 A (TMEM86A), a putative lysoplasmalogenase. Untargeted lipidomic analysis demonstrates that adipocyte-specific TMEM86A-knockout (AKO) increases lysoplasmalogen content in adipose tissue, including plasmenyl lysophosphatidylethanolamine 18:0 (LPE P-18:0). Surprisingly, TMEM86A AKO increases protein kinase A signalling pathways owing to inhibition of phosphodiesterase 3B and elevation of cyclic adenosine monophosphate. TMEM86A AKO upregulates mitochondrial oxidative metabolism, elevates energy expenditure, and protects mice from metabolic dysfunction induced by high-fat feeding. Importantly, the effects of TMEM86A AKO are largely reproduced in vitro and in vivo by LPE P-18:0 supplementation. LPE P-18:0 levels are significantly lower in adipose tissue of human patients with obesity, suggesting that TMEM86A inhibition or lysoplasmalogen supplementation might be therapeutic approaches for preventing or treating obesity-related metabolic diseases.
Collapse
|
26
|
Takic M, Pokimica B, Petrovic-Oggiano G, Popovic T. Effects of Dietary α-Linolenic Acid Treatment and the Efficiency of Its Conversion to Eicosapentaenoic and Docosahexaenoic Acids in Obesity and Related Diseases. Molecules 2022; 27:molecules27144471. [PMID: 35889342 PMCID: PMC9317994 DOI: 10.3390/molecules27144471] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/06/2023] Open
Abstract
The essential fatty acid alpha-linolenic acid (ALA) is present in high amounts in oils such as flaxseed, soy, hemp, rapeseed, chia, and perilla, while stearidonic acid is abundant in echium oil. ALA is metabolized to eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) by desaturases and elongases in humans. The conversion of ALA to EPA and DHA is limited, and these long-chain n−3 polyunsaturated fatty acids (PUFAs) are mainly provided from dietary sources (fish and seafood). This review provides an overview of studies that explored the effects of dietary supplementation with ALA in obesity and related diseases. The obesity-associated changes of desaturase and elongase activities are summarized, as they could influence the metabolic conversion of ALA. Generally, supplementation with ALA or ALA-rich oils leads to an increase in EPA levels and has no effect on DHA or omega-3 index. According to the literature data, stearidonic acid could enhance conversion of ALA to long-chain n−3 PUFA in obesity. Recent studies confirm that EPA and DHA intake should be considered as a primary dietary treatment strategy for improving the omega-3 index in obesity and related diseases.
Collapse
|
27
|
Motahari-Rad H, Subiri A, Soler R, Ocaña L, Alcaide J, Rodríguez-Capitan J, Buil V, el Azzouzi H, Ortega-Gomez A, Bernal-Lopez R, Insenser M, Tinahones FJ, Murri M. The Effect of Sex and Obesity on the Gene Expression of Lipid Flippases in Adipose Tissue. J Clin Med 2022; 11:jcm11133878. [PMID: 35807162 PMCID: PMC9267438 DOI: 10.3390/jcm11133878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 01/25/2023] Open
Abstract
Molecular mechanisms behind obesity and sex-related effects in adipose tissue remain elusive. During adipocyte expansion, adipocytes undergo drastic remodelling of lipid membrane compositions. Lipid flippases catalyse phospholipid translocation from exoplasmic to the cytoplasmic leaflet of membranes. The present study aimed to analyse the effect of sex, obesity, and their interactions on the gene expression of two lipid flippases—ATP8A1 and ATP8B1—and their possible microRNA (miR) modulators in visceral adipose tissue (VAT). In total, 12 normal-weight subjects (5 premenopausal women and 7 men) and 13 morbidly obese patients (7 premenopausal women and 6 men) were submitted to surgery, and VAT samples were obtained. Gene expression levels of ATP8A1, ATP8B1, miR-548b-5p, and miR-4643 were measured in VAT. Our results showed a marked influence of obesity on VAT ATP8A1 and ATP8B1, although the effects of obesity were stronger in men for ATP8A1. Both genes positively correlated with obesity and metabolic markers. Furthermore, ATP8B1 was positively associated with miR-548b-5p and negatively associated with miR-4643. Both miRs were also affected by sex. Thus, lipid flippases are altered by obesity in VAT in a sex-specific manner. Our study provides a better understanding of the sex-specific molecular mechanisms underlying obesity, which may contribute to the development of sex-based precision medicine.
Collapse
Affiliation(s)
- Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran;
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
| | - Alba Subiri
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Rocio Soler
- Clinical Management Unit (UGC) of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (R.S.); (L.O.)
| | - Luis Ocaña
- Clinical Management Unit (UGC) of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain; (R.S.); (L.O.)
| | - Juan Alcaide
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Jorge Rodríguez-Capitan
- Clinical Management Unit (UGC) of Heart, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 29010 Málaga, Spain;
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Veronica Buil
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Hamid el Azzouzi
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Almudena Ortega-Gomez
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
| | - Rosa Bernal-Lopez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Clinical Management Unit (UGC) of Internal Medicine, IBIMA, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain
- Correspondence: (M.I.); (M.M.)
| | - Francisco J. Tinahones
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Faculty of Medicine, University of Malaga, 29010 Malaga, Spain;
| | - Mora Murri
- Clinical Management Unit (UGC) of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain; (A.S.); (J.A.); (A.O.-G.); (F.J.T.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIlBEROBN), Instituto de Salud Carlos III, 29010 Málaga, Spain;
- Correspondence: (M.I.); (M.M.)
| |
Collapse
|
28
|
Sailer S, Lackner K, Pras-Raves ML, Wever EJ, van Klinken JB, Dane AD, Geley S, Koch J, Golderer G, Werner-Felmayer G, Keller MA, Zwerschke W, Vaz FM, Werner ER, Watschinger K. Adaptations of the 3T3-L1 adipocyte lipidome to defective ether lipid catabolism upon alkylglycerol monooxygenase knockdown. J Lipid Res 2022; 63:100222. [PMID: 35537527 PMCID: PMC9192799 DOI: 10.1016/j.jlr.2022.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 10/24/2022] Open
|
29
|
Ku EJ, Kim BR, Lee JI, Lee YK, Oh TJ, Jang HC, Choi SH. The Anti-Atherosclerosis Effect of Anakinra, a Recombinant Human Interleukin-1 Receptor Antagonist, in Apolipoprotein E Knockout Mice. Int J Mol Sci 2022; 23:ijms23094906. [PMID: 35563294 PMCID: PMC9104865 DOI: 10.3390/ijms23094906] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/10/2022] Open
Abstract
Interleukin (IL)-1β plays an important role in atherosclerosis pathogenesis. We aimed to investigate the effect of anakinra, a recombinant human IL-1 receptor antagonist, on the progression of atherosclerosis in apolipoprotein E knockout (ApoE−/−) mice. ApoE−/− mice (8-week male) were treated with saline (control), anakinra 10, 25, and 50 mg/kg, respectively (n = 10 in each group). Mice were fed a standard chow (4 weeks) followed by an atherogenic diet (35kcal% fat, 1.25% cholesterol, 12 weeks). Atheromatous plaques in ApoE−/− mice and the expression of inflammatory genes and signaling pathways in human umbilical vein endothelial cells (HUVECs), rat aortic smooth muscle cells (RAOSMCs), and 3T3-L1 adipocytes were assessed. Anakinra reduced the plaque size of the aortic arch (30.6% and 25.2% at the 25 mg/kg and 50 mg/kg doses, both p < 0.05) and serum triglyceride in ApoE−/− mice and suppressed inflammatory genes (IL-1β and IL-6) expressions in HUVECs and RAOSMCs (all p < 0.05). In RAOSMCs, anakinra reduced metalloproteinase-9 expression in a dose-dependent manner and inhibited cell migration. Anakinra-treated mice exhibited trends of lower CD68+ macrophage infiltration in visceral fat and monocyte chemoattractant protein-1 expression was reduced in 3T3-L1 adipocytes. Anakinra could be a useful component for complementary treatment with a standard regimen to reduce the residual cardiovascular risk.
Collapse
Affiliation(s)
- Eu Jeong Ku
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea;
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Bo-Rahm Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Jee-In Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Yun Kyung Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hak C. Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-31-787-7033
| |
Collapse
|
30
|
Wang Q, Sun N, Kunzke T, Buck A, Shen J, Prade VM, Stöckl B, Wang J, Feuchtinger A, Walch A. A simple preparation step to remove excess liquid lipids in white adipose tissue enabling improved detection of metabolites via MALDI-FTICR imaging MS. Histochem Cell Biol 2022; 157:595-605. [PMID: 35391562 PMCID: PMC9114030 DOI: 10.1007/s00418-022-02088-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 11/10/2022]
Abstract
Matrix-assisted laser desorption ionization (MALDI) Fourier transform ion cyclotron resonance (FTICR) imaging mass spectrometry (MS) is a powerful technology used to analyze metabolites in various tissues. However, it faces significant challenges in studying adipose tissues. Poor matrix distribution and crystallization caused by excess liquid lipids on the surface of tissue sections hamper m/z species detection, an adverse effect that particularly presents in lipid-rich white adipose tissue (WAT). In this study, we integrated a simple and low-cost preparation step into the existing MALDI-FTICR imaging MS pipeline. The new method—referred to as filter paper application—is characterized by an easy sample handling and high reproducibility. The aforementioned filter paper is placed onto the tissue prior to matrix application in order to remove the layer of excess liquid lipids. Consequently, MALDI-FTICR imaging MS detection was significantly improved, resulting in a higher number of detected m/z species and higher ion intensities. After analyzing various durations of filter paper application, 30 s was found to be optimal, resulting in the detection of more than 3700 m/z species. Apart from the most common lipids found in WAT, other molecules involved in various metabolic pathways were detected, including nucleotides, carbohydrates, and amino acids. Our study is the first to propose a solution to a specific limitation of MALDI-FTICR imaging MS in investigating lipid-rich WAT. The filter paper approach can be performed quickly and is particularly effective for achieving uniform matrix distribution on fresh frozen WAT while maintaining tissue integrity. It thus helps to gain insight into the metabolism in WAT.
Collapse
Affiliation(s)
- Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Achim Buck
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Jian Shen
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Verena M Prade
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Barbara Stöckl
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Jun Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764, Neuherberg, Germany.
| |
Collapse
|
31
|
Schooneveldt YL, Paul S, Calkin AC, Meikle PJ. Ether Lipids in Obesity: From Cells to Population Studies. Front Physiol 2022; 13:841278. [PMID: 35309067 PMCID: PMC8927733 DOI: 10.3389/fphys.2022.841278] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Ether lipids are a unique class of glycero- and glycerophospho-lipid that carry an ether or vinyl ether linked fatty alcohol at the sn-1 position of the glycerol backbone. These specialised lipids are important endogenous anti-oxidants with additional roles in regulating membrane fluidity and dynamics, intracellular signalling, immunomodulation and cholesterol metabolism. Lipidomic profiling of human population cohorts has identified new associations between reduced circulatory plasmalogen levels, an abundant and biologically active sub-class of ether lipids, with obesity and body-mass index. These findings align with the growing body of work exploring novel roles for ether lipids within adipose tissue. In this regard, ether lipids have now been linked to facilitating lipid droplet formation, regulating thermogenesis and mediating beiging of white adipose tissue in early life. This review will assess recent findings in both population studies and studies using cell and animal models to delineate the functional and protective roles of ether lipids in the setting of obesity. We will also discuss the therapeutic potential of ether lipid supplementation to attenuate diet-induced obesity.
Collapse
Affiliation(s)
- Yvette L. Schooneveldt
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Sudip Paul
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| | - Anna C. Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Anna C. Calkin,
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
- Peter J. Meikle,
| |
Collapse
|
32
|
Mocciaro G, Gastaldelli A. Obesity-Related Insulin Resistance: The Central Role of Adipose Tissue Dysfunction. Handb Exp Pharmacol 2022; 274:145-164. [PMID: 35192055 DOI: 10.1007/164_2021_573] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Obesity is a key player in the onset and progression of insulin resistance (IR), a state by which insulin-sensitive cells fail to adequately respond to insulin action. IR is a reversible condition, but if untreated leads to type 2 diabetes alongside increasing cardiovascular risk. The link between obesity and IR has been widely investigated; however, some aspects are still not fully characterized.In this chapter, we introduce key aspects of the pathophysiology of IR and its intimate connection with obesity. Specifically, we focus on the role of adipose tissue dysfunction (quantity, quality, and distribution) as a driver of whole-body IR. Furthermore, we discuss the obesity-related lipidomic remodeling occurring in adipose tissue, liver, and skeletal muscle. Key mechanisms linking lipotoxicity to IR in different tissues and metabolic alterations (i.e., fatty liver and diabetes) and the effect of weight loss on IR are also reported while highlighting knowledge gaps.
Collapse
Affiliation(s)
- Gabriele Mocciaro
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy.
| |
Collapse
|
33
|
Khanna D, Khanna S, Khanna P, Kahar P, Patel BM. Obesity: A Chronic Low-Grade Inflammation and Its Markers. Cureus 2022; 14:e22711. [PMID: 35386146 PMCID: PMC8967417 DOI: 10.7759/cureus.22711] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
As the prevalence of obesity continues to rise, the world is facing a major public health concern. Obesity is a complex disease associated with an increase in several inflammatory markers, leading to chronic low-grade inflammation. Of multifactorial etiology, it is often used as a measurement of morbidity and mortality. There remains much unknown regarding the association between obesity and inflammation. This review seeks to compile scientific literature on obesity and its associated inflammatory markers in chronic disease and further discusses the role of adipose tissue, macrophages, B-cells, T-cells, fatty acids, amino acids, adipokines, and hormones in obesity. Data were obtained using PubMed and Google Scholar. Obesity, inflammation, immune cells, hormones, fatty acids, and others were search words used to acquire relevant articles. Studies suggest brown adipose tissue is negatively associated with body mass index (BMI) and body fat percentage. Researchers also found the adipose tissue of lean individuals predominantly secretes anti-inflammatory markers, while in obese individuals more pro-inflammatory markers are secreted. Many studies found that adipose tissue in obese individuals showed a shift in immune cells from anti-inflammatory M2 macrophages to pro-inflammatory M1 macrophages, which was also correlated with insulin resistance. Obese individuals generally present with higher levels of hormones such as leptin, visfatin, and resistin. With obesity on the rise globally, it is predicted that severe obesity will become most common amongst low-income adults, black individuals, and women by 2030, making the need for intervention urgent. Further investigation into the association between obesity and inflammation is required to understand the mechanism behind this disease.
Collapse
Affiliation(s)
- Deepesh Khanna
- Foundational Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Siya Khanna
- Foundational Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Pragya Khanna
- Pediatrics, Gujarat Medical Education and Research Society (GMERS) Medical College, Vadnagar, IND
| | - Payal Kahar
- Department of Health Sciences, Florida Gulf Coast University, Fort Myers, USA
| | - Bhavesh M Patel
- Pediatrics, Gujarat Medical Education and Research Society (GMERS) Medical College, Vadnagar, IND
| |
Collapse
|
34
|
Oliveira AA, Róg T, da Silva ABF, Amaro RE, Johnson MS, Postila PA. Examining the Effect of Charged Lipids on Mitochondrial Outer Membrane Dynamics Using Atomistic Simulations. Biomolecules 2022; 12:183. [PMID: 35204684 PMCID: PMC8961577 DOI: 10.3390/biom12020183] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/11/2022] Open
Abstract
The outer mitochondrial membrane (OMM) is involved in multiple cellular functions such as apoptosis, inflammation and signaling via its membrane-associated and -embedded proteins. Despite the central role of the OMM in these vital phenomena, the structure and dynamics of the membrane have regularly been investigated in silico using simple two-component models. Accordingly, the aim was to generate the realistic multi-component model of the OMM and inspect its properties using atomistic molecular dynamics (MD) simulations. All major lipid components, phosphatidylinositol (PI), phosphatidylcholine (PC), phosphatidylethanolamine (PE), and phosphatidylserine (PS), were included in the probed OMM models. Because increased levels of anionic PS lipids have potential effects on schizophrenia and, more specifically, on monoamine oxidase B enzyme activity, the effect of varying the PS concentration was explored. The MD simulations indicate that the complex membrane lipid composition (MLC) behavior is notably different from the two-component PC-PE model. The MLC changes caused relatively minor effects on the membrane structural properties such as membrane thickness or area per lipid; however, notable effects could be seen with the dynamical parameters at the water-membrane interface. Increase of PS levels appears to slow down lateral diffusion of all lipids and, in general, the presence of anionic lipids reduced hydration and slowed down the PE headgroup rotation. In addition, sodium ions could neutralize the membrane surface, when PI was the main anionic component; however, a similar effect was not seen for high PS levels. Based on these results, it is advisable for future studies on the OMM and its protein or ligand partners, especially when wanting to replicate the correct properties on the water-membrane interface, to use models that are sufficiently complex, containing anionic lipid types, PI in particular.
Collapse
Affiliation(s)
- Aline A. Oliveira
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA 92093-0340, USA; (A.A.O.); (R.E.A.)
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, São Carlos 13560-970, Brazil;
| | - Tomasz Róg
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland;
| | - Albérico B. F. da Silva
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, São Carlos 13560-970, Brazil;
| | - Rommie E. Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA 92093-0340, USA; (A.A.O.); (R.E.A.)
| | - Mark S. Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- InFLAMES Research Flagship Center, Åbo Akademi University, 20520 Turku, Finland
| | - Pekka A. Postila
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA 92093-0340, USA; (A.A.O.); (R.E.A.)
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- InFLAMES Research Flagship Center, Åbo Akademi University, 20520 Turku, Finland
- Institute of Biomedicine, Kiinamyllynkatu 10, Integrative Physiology and Pharmacy, University of Turku, FI-20520 Turku, Finland
- Aurlide Ltd., FI-21420 Lieto, Finland
- InFLAMES Research Flagship Center, University of Turku, FI-20520 Turku, Finland
| |
Collapse
|
35
|
Oates EH, Antoniewicz MR. Coordinated reprogramming of metabolism and cell function in adipocytes from proliferation to differentiation. Metab Eng 2021; 69:221-230. [PMID: 34929419 DOI: 10.1016/j.ymben.2021.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/25/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
Adipose tissue plays a major role in regulating lipid and energy homeostasis by storing excess nutrients, releasing energetic substrates through lipolysis, and regulating metabolism of other tissues and organs through endocrine and paracrine signaling. Adipocytes within fat tissues store excess nutrients through increased cell number (hyperplasia), increased cell size (hypertrophy), or both. The differentiation of pre-adipocytes into mature lipid-accumulating adipocytes requires a complex interaction of metabolic pathways that is still incompletely understood. Here, we applied parallel labeling experiments and 13C-metabolic flux analysis to quantify precise metabolic fluxes in proliferating and differentiated 3T3-L1 cells, a widely used model to study adipogenesis. We found that morphological and biomass composition changes in adipocytes were accompanied by significant shifts in metabolic fluxes, encompassing all major metabolic pathways. In contrast to proliferating cells, differentiated adipocytes 1) increased glucose uptake and redirected glucose utilization from lactate production to lipogenesis and energy generation; 2) increased pathway fluxes through glycolysis, oxidative pentose phosphate pathway and citric acid cycle; 3) reduced lactate secretion, resulting in increased ATP generation via oxidative phosphorylation; 4) rewired glutamine metabolism, from glutaminolysis to de novo glutamine synthesis; 5) increased cytosolic NADPH production, driven mostly by increased cytosolic malic enzyme flux; 6) increased production of monounsaturated C16:1; and 7) activated a mitochondrial pyruvate cycle through simultaneous activity of pyruvate carboxylase, malate dehydrogenase and malic enzyme. Taken together, these results quantitatively highlight the complex interplay between pathway fluxes and cell function in adipocytes, and suggest a functional role for metabolic reprogramming in adipose differentiation and lipogenesis.
Collapse
Affiliation(s)
- Eleanor H Oates
- Department of Chemical and Biomolecular Engineering, Metabolic Engineering and Systems Biology Laboratory, University of Delaware, Newark, DE, 19716, USA
| | - Maciek R Antoniewicz
- Department of Chemical and Biomolecular Engineering, Metabolic Engineering and Systems Biology Laboratory, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
36
|
Palavicini JP, Chavez-Velazquez A, Fourcaudot M, Tripathy D, Pan M, Norton L, DeFronzo RA, Shannon CE. The Insulin-Sensitizer Pioglitazone Remodels Adipose Tissue Phospholipids in Humans. Front Physiol 2021; 12:784391. [PMID: 34925073 PMCID: PMC8674727 DOI: 10.3389/fphys.2021.784391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-sensitizer pioglitazone exerts its cardiometabolic benefits in type 2 diabetes (T2D) through a redistribution of body fat, from ectopic and visceral areas to subcutaneous adipose depots. Whereas excessive weight gain and lipid storage in obesity promotes insulin resistance and chronic inflammation, the expansion of subcutaneous adipose by pioglitazone is associated with a reversal of these immunometabolic deficits. The precise events driving this beneficial remodeling of adipose tissue with pioglitazone remain unclear, and whether insulin-sensitizers alter the lipidomic composition of human adipose has not previously been investigated. Using shotgun lipidomics, we explored the molecular lipid responses in subcutaneous adipose tissue following 6months of pioglitazone treatment (45mg/day) in obese humans with T2D. Despite an expected increase in body weight following pioglitazone treatment, no robust effects were observed on the composition of storage lipids (i.e., triglycerides) or the content of lipotoxic lipid species (e.g., ceramides and diacylglycerides) in adipose tissue. Instead, pioglitazone caused a selective remodeling of the glycerophospholipid pool, characterized by a decrease in lipids enriched for arachidonic acid, such as plasmanylethanolamines and phosphatidylinositols. This contributed to a greater overall saturation and shortened chain length of fatty acyl groups within cell membrane lipids, changes that are consistent with the purported induction of adipogenesis by pioglitazone. The mechanism through which pioglitazone lowered adipose tissue arachidonic acid, a major modulator of inflammatory pathways, did not involve alterations in phospholipase gene expression but was associated with a reduction in its precursor linoleic acid, an effect that was also observed in skeletal muscle samples from the same subjects. These findings offer important insights into the biological mechanisms through which pioglitazone protects the immunometabolic health of adipocytes in the face of increased lipid storage.
Collapse
Affiliation(s)
- Juan P. Palavicini
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alberto Chavez-Velazquez
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Marcel Fourcaudot
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Meixia Pan
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Luke Norton
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ralph A. DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Christopher E. Shannon
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
37
|
Bozelli JC, Azher S, Epand RM. Plasmalogens and Chronic Inflammatory Diseases. Front Physiol 2021; 12:730829. [PMID: 34744771 PMCID: PMC8566352 DOI: 10.3389/fphys.2021.730829] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022] Open
Abstract
It is becoming widely acknowledged that lipids play key roles in cellular function, regulating a variety of biological processes. Lately, a subclass of glycerophospholipids, namely plasmalogens, has received increased attention due to their association with several degenerative and metabolic disorders as well as aging. All these pathophysiological conditions involve chronic inflammatory processes, which have been linked with decreased levels of plasmalogens. Currently, there is a lack of full understanding of the molecular mechanisms governing the association of plasmalogens with inflammation. However, it has been shown that in inflammatory processes, plasmalogens could trigger either an anti- or pro-inflammation response. While the anti-inflammatory response seems to be linked to the entire plasmalogen molecule, its pro-inflammatory response seems to be associated with plasmalogen hydrolysis, i.e., the release of arachidonic acid, which, in turn, serves as a precursor to produce pro-inflammatory lipid mediators. Moreover, as plasmalogens comprise a large fraction of the total lipids in humans, changes in their levels have been shown to change membrane properties and, therefore, signaling pathways involved in the inflammatory cascade. Restoring plasmalogen levels by use of plasmalogen replacement therapy has been shown to be a successful anti-inflammatory strategy as well as ameliorating several pathological hallmarks of these diseases. The purpose of this review is to highlight the emerging role of plasmalogens in chronic inflammatory disorders as well as the promising role of plasmalogen replacement therapy in the treatment of these pathologies.
Collapse
Affiliation(s)
- José Carlos Bozelli
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON, Canada
| | - Sayed Azher
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, Health Sciences Centre, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
38
|
Shinde AB, Zaganjor E. AdipoAtlas: Mapping out human white adipose tissue. Cell Rep Med 2021; 2:100429. [PMID: 34755140 PMCID: PMC8561306 DOI: 10.1016/j.xcrm.2021.100429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In this issue of Cell Reports Medicine, Lange and colleagues1 significantly improve lipid identification accuracy, detection, and quantification to provide AdipoAtlas, an in-depth lipidomic profile of human white adipose tissue (WAT). Importantly, they define obesity-mediated lipid alterations, which may provide insight into the etiology of associated diseases.
Collapse
Affiliation(s)
- Abhijit B. Shinde
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232-0615, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232-0615, USA
| |
Collapse
|
39
|
Lange M, Angelidou G, Ni Z, Criscuolo A, Schiller J, Blüher M, Fedorova M. AdipoAtlas: A reference lipidome for human white adipose tissue. Cell Rep Med 2021; 2:100407. [PMID: 34755127 PMCID: PMC8561168 DOI: 10.1016/j.xcrm.2021.100407] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/29/2021] [Accepted: 08/26/2021] [Indexed: 01/16/2023]
Abstract
Obesity, characterized by expansion and metabolic dysregulation of white adipose tissue (WAT), has reached pandemic proportions and acts as a primer for a wide range of metabolic disorders. Remodeling of WAT lipidome in obesity and associated comorbidities can explain disease etiology and provide valuable diagnostic and prognostic markers. To support understanding of WAT lipidome remodeling at the molecular level, we provide in-depth lipidomics profiling of human subcutaneous and visceral WAT of lean and obese individuals. We generate a human WAT reference lipidome by performing tissue-tailored preanalytical and analytical workflows, which allow accurate identification and semi-absolute quantification of 1,636 and 737 lipid molecular species, respectively. Deep lipidomic profiling allows identification of main lipid (sub)classes undergoing depot-/phenotype-specific remodeling. Previously unanticipated diversity of WAT ceramides is now uncovered. AdipoAtlas reference lipidome serves as a data-rich resource for the development of WAT-specific high-throughput methods and as a scaffold for systems medicine data integration.
Collapse
Affiliation(s)
- Mike Lange
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Georgia Angelidou
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Angela Criscuolo
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
- Thermo Fisher Scientific, Dreieich, Germany
| | - Jürgen Schiller
- Institute of Medical Physics and Biophysics, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
40
|
Mayneris-Perxachs J, Meikle P, Mousa A, Naderpoor N, Fernández-Real JM, de Courten B. Novel Relationship Between Plasmalogen Lipid Signatures and Carnosine in Humans. Mol Nutr Food Res 2021; 65:e2100164. [PMID: 34328693 DOI: 10.1002/mnfr.202100164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/25/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Carnosine is a naturally occurring dipeptide abundant in the skeletal and cardiac muscle and brain, which has been shown to improve glucose metabolism and cardiovascular risk. This study showed that carnosine supplementation had positive changes on plasma lipidome. Here, this study aimed to establish the relationship of muscle carnosine and serum carnosinase-1 with cardiometabolic risk factors and the lipidome. METHODS AND RESULTS This study profiles >450 lipid species in 65 overweight/obese nondiabetic individuals. Intensive metabolic testing is conducted using direct gold-standard measures of adiposity, insulin sensitivity and secretion, as well as measurement of serum inflammatory cytokines and adipokines. Muscle carnosine is negatively associated with 2-h glucose concentrations, whereas serum carnosinase-1 levels are negatively associated with insulin sensitivity and positively with IL-18. O-PLS and machine learning analyses reveal a strong association of muscle carnosine with ether lipids, particularly arachidonic acid-containing plasmalogens. Carnosinase-1 levels are positively associated with total phosphatidylethanolamines, but negatively with lysoalkylphosphatidylcholines, trihexosylceramides, and gangliosides. In particular, alkylphosphatidylethanolamine species containing arachidonic acid are positively associated with carnosinase-1. CONCLUSION These associations reinforce the role of muscle carnosine and serum carnosinase-1 in the interplay among low-grade chronic inflammation, glucose homeostasis, and insulin sensitivity.
Collapse
Affiliation(s)
- Jordi Mayneris-Perxachs
- Department of Endocrinology, Diabetes and Nutrition, Hospital of Girona "Dr Josep Trueta," University of Girona, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,CIBERobn Pathophysiology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain
| | - Peter Meikle
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, Australia
| | - Negar Naderpoor
- Monash Centre for Health Research and Implementation, Monash University, Melbourne, Australia
| | - José Manuel Fernández-Real
- Department of Endocrinology, Diabetes and Nutrition, Hospital of Girona "Dr Josep Trueta," University of Girona, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,CIBERobn Pathophysiology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain.,Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Barbora de Courten
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
41
|
Kyle JE, Aimo L, Bridge AJ, Clair G, Fedorova M, Helms JB, Molenaar MR, Ni Z, Orešič M, Slenter D, Willighagen E, Webb-Robertson BJM. Interpreting the lipidome: bioinformatic approaches to embrace the complexity. Metabolomics 2021; 17:55. [PMID: 34091802 DOI: 10.1007/s11306-021-01802-6] [Citation(s) in RCA: 4] [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: 02/10/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Improvements in mass spectrometry (MS) technologies coupled with bioinformatics developments have allowed considerable advancement in the measurement and interpretation of lipidomics data in recent years. Since research areas employing lipidomics are rapidly increasing, there is a great need for bioinformatic tools that capture and utilize the complexity of the data. Currently, the diversity and complexity within the lipidome is often concealed by summing over or averaging individual lipids up to (sub)class-based descriptors, losing valuable information about biological function and interactions with other distinct lipids molecules, proteins and/or metabolites. AIM OF REVIEW To address this gap in knowledge, novel bioinformatics methods are needed to improve identification, quantification, integration and interpretation of lipidomics data. The purpose of this mini-review is to summarize exemplary methods to explore the complexity of the lipidome. KEY SCIENTIFIC CONCEPTS OF REVIEW Here we describe six approaches that capture three core focus areas for lipidomics: (1) lipidome annotation including a resolvable database identifier, (2) interpretation via pathway- and enrichment-based methods, and (3) understanding complex interactions to emphasize specific steps in the analytical process and highlight challenges in analyses associated with the complexity of lipidome data.
Collapse
Affiliation(s)
- Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Lucila Aimo
- Swiss-Prot Group, SIB Swiss Institute of Bioinformatics, 1 rue Michel-Servet, 1211, Geneva 4, Switzerland
| | - Alan J Bridge
- Swiss-Prot Group, SIB Swiss Institute of Bioinformatics, 1 rue Michel-Servet, 1211, Geneva 4, Switzerland
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Center for Biotechnology and Biomedicine, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany
| | - J Bernd Helms
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martijn R Molenaar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Center for Biotechnology and Biomedicine, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany
| | - Matej Orešič
- School of Medical Sciences, Örebro University, 702 81, Örebro, Sweden
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Denise Slenter
- Department of Bioinformatics-BiGCaT, NUTRIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Egon Willighagen
- Department of Bioinformatics-BiGCaT, NUTRIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
| | | |
Collapse
|
42
|
Paavola T, Bergmann U, Kuusisto S, Kakko S, Savolainen MJ, Salonurmi T. Distinct Fatty Acid Compositions of HDL Phospholipids Are Characteristic of Metabolic Syndrome and Premature Coronary Heart Disease-Family Study. Int J Mol Sci 2021; 22:ijms22094908. [PMID: 34066314 PMCID: PMC8124224 DOI: 10.3390/ijms22094908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 02/07/2023] Open
Abstract
HDL particles can be structurally modified in atherosclerotic disorders associated with low HDL cholesterol level (HDL-C). We studied whether the lipidome of the main phosphatidylcholine (PC), lysophosphatidylcholine (LPC) and sphingomyelin (SM) species of HDL2 and HDL3 subfractions is associated with premature coronary heart disease (CHD) or metabolic syndrome (MetS) in families where common low HDL-C predisposes to premature CHD. The lipidome was analyzed by LC-MS. Lysophosphatidylcholines were depleted of linoleic acid relative to more saturated and shorter-chained acids containing species in MetS compared with non-affected subjects: the ratio of palmitic to linoleic acid was elevated by more than 30%. A minor PC (16:0/16:1) was elevated (28–40%) in MetS. The contents of oleic acid containing PCs were elevated relative to linoleic acid containing PCs in MetS; the ratio of PC (16:0/18:1) to PC (16:0/18:2) was elevated by 11–16%. Certain PC and SM ratios, e.g., PC (18:0/20:3) to PC (16:0/18:2) and a minor SM 36:2 to an abundant SM 34:1, were higher (11–36%) in MetS and CHD. The fatty acid composition of certain LPCs and PCs displayed a characteristic pattern in MetS, enriched with palmitic, palmitoleic or oleic acids relative to linoleic acid. Certain PC and SM ratios related consistently to CHD and MetS.
Collapse
Affiliation(s)
- Timo Paavola
- Research Center for Internal Medicine, Department of Internal Medicine, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
| | - Ulrich Bergmann
- Protein Analysis Core Facility, Biocenter Oulu, University of Oulu, 90570 Oulu, Finland
| | - Sanna Kuusisto
- Computational Medicine, Faculty of Medicine, Biocenter Oulu, University of Oulu, 90570 Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland
| | - Sakari Kakko
- Research Center for Internal Medicine, Department of Internal Medicine, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
| | - Markku J Savolainen
- Research Center for Internal Medicine, Department of Internal Medicine, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
| | - Tuire Salonurmi
- Research Center for Internal Medicine, Department of Internal Medicine, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90200 Oulu, Finland
| |
Collapse
|
43
|
Shetty SS, Kumari S. Fatty acids and their role in type-2 diabetes (Review). Exp Ther Med 2021; 22:706. [PMID: 34007315 PMCID: PMC8120551 DOI: 10.3892/etm.2021.10138] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Age, lifestyle and diet are major risk factors for the onset of type 2 diabetes mellitus (T2DM). Insulin resistance (IR) and β-cell dysfunction underlie the pathophysiology of T2DM. Diabetic populations are also prone to lipid and lipoprotein abnormalities as an indirect effect of IR on key metabolic enzymes. However, recent studies suggested that lipid changes may not only be a consequence of impaired glucose metabolism but also a causative factor. Fatty acids (FAs) influence translocation of glucose transporters and insulin receptor binding and signalling, in addition to cell membrane fluidity and permeability. It is thus suggested that FAs may have an essential role in the development of IR and T2DM. Specific combinations of FAs within phospholipids and triglycerides were indicated to exhibit the strongest associations with the risk of T2DM. The aim of the present review was to investigate the role of FAs in the pathogenesis of T2DM, as it has yet to be fully elucidated.
Collapse
Affiliation(s)
- Shilpa S Shetty
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - Suchetha Kumari
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| |
Collapse
|
44
|
AGMO Inhibitor Reduces 3T3-L1 Adipogenesis. Cells 2021; 10:cells10051081. [PMID: 34062826 PMCID: PMC8147360 DOI: 10.3390/cells10051081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Alkylglycerol monooxygenase (AGMO) is a tetrahydrobiopterin (BH4)-dependent enzyme with major expression in the liver and white adipose tissue that cleaves alkyl ether glycerolipids. The present study describes the disclosure and biological characterization of a candidate compound (Cp6), which inhibits AGMO with an IC50 of 30–100 µM and 5–20-fold preference of AGMO relative to other BH4-dependent enzymes, i.e., phenylalanine-hydroxylase and nitric oxide synthase. The viability and metabolic activity of mouse 3T3-L1 fibroblasts, HepG2 human hepatocytes and mouse RAW264.7 macrophages were not affected up to 10-fold of the IC50. However, Cp6 reversibly inhibited the differentiation of 3T3-L1 cells towards adipocytes, in which AGMO expression was upregulated upon differentiation. Cp6 reduced the accumulation of lipid droplets in adipocytes upon differentiation and in HepG2 cells exposed to free fatty acids. Cp6 also inhibited IL-4-driven differentiation of RAW264.7 macrophages towards M2-like macrophages, which serve as adipocyte progenitors in adipose tissue. Collectively, the data suggest that pharmacologic AGMO inhibition may affect lipid storage.
Collapse
|
45
|
Shao J, Pan T, Wang J, Tang T, Li Y, Jia X, Lai S. Integrated Proteomics and Metabolomics Analysis of Perirenal Adipose Tissue in Obese Rabbits Treated with a Restricted Diet. BIOLOGY 2021; 10:321. [PMID: 33921318 PMCID: PMC8069198 DOI: 10.3390/biology10040321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
In recent years, many people have shown an excess of fat accumulation. Known as obesity, this lesion poses an increased risk for multiple diseases, such as endocrine disease, diabetes, and cancer, and has reached epidemic proportions. Accompanied by the development of obesity, concern over body image and weight loss behavior is a growing social problem and public health threat, causing concern for many health professionals. However, the consequences of rapid weight loss remain largely unclear. Here, we applied an integrated proteomics and metabolomics analysis to investigate the effects of dieting on the proteins and metabolites in obese rabbits. Our study revealed that 343 differentially expressed proteins (136 upregulated and 207 downregulated) and 150 differentially expressed metabolites (91 upregulated and 59 downregulated) were identified. These molecules are mainly involved in the biological processes, including amino acid metabolism, lipid metabolism, and membrane and cytoskeleton reconstruction. The integrated analysis found that mevalonic acid, arachidonic acid, 15(S)-HpETE, cholecalciferol, hydrocortisone, lipoxin B4, lithocholic acid, etc. were associated with multiple pathways, and they may be the key factors to fight inflammation induced by a high-fat diet (HFD). Overall, this study provides further insight into the consequences of dieting-mediated weight loss and may contribute to the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| | - Ting Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China;
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| | - Tao Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| | - Yanhong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (J.S.); (J.W.); (T.T.); (Y.L.); (X.J.)
| |
Collapse
|
46
|
Palau-Rodriguez M, Marco-Ramell A, Casas-Agustench P, Tulipani S, Miñarro A, Sanchez-Pla A, Murri M, Tinahones FJ, Andres-Lacueva C. Visceral Adipose Tissue Phospholipid Signature of Insulin Sensitivity and Obesity. J Proteome Res 2021; 20:2410-2419. [PMID: 33760621 PMCID: PMC8631729 DOI: 10.1021/acs.jproteome.0c00918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Alterations in visceral adipose tissue
(VAT) are closely linked
to cardiometabolic abnormalities. The aim of this work is to define
a metabolic signature in VAT of insulin resistance (IR) dependent
on, and independent of, obesity. An untargeted UPLC-Q-Exactive metabolomic
approach was carried out on the VAT of obese insulin-sensitive (IS)
and insulin-resistant subjects (N = 11 and N = 25, respectively) and nonobese IS and IR subjects (N = 25 and N = 10, respectively). The VAT
metabolome in obesity was defined among other things by changes in
the metabolism of lipids, nucleotides, carbohydrates, and amino acids,
whereas when combined with high IR, it affected the metabolism of
18 carbon fatty acyl-containing phospholipid species. A multimetabolite
model created by glycerophosphatidylinositol (18:0); glycerophosphatidylethanolamine
(18:2); glycerophosphatidylserine (18:0); and glycerophosphatidylcholine
(18:0/18:1), (18:2/18:2), and (18:2/18:3) exhibited a highly predictive
performance to identify the metabotype of “insulin-sensitive
obesity” among obese individuals [area under the curve (AUC)
96.7% (91.9–100)] and within the entire study population [AUC
87.6% (79.0–96.2)]. We demonstrated that IR has a unique and
shared metabolic signature dependent on, and independent of, obesity.
For it to be used in clinical practice, these findings need to be
validated in a more accessible sample, such as blood.
Collapse
Affiliation(s)
- Magalí Palau-Rodriguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Anna Marco-Ramell
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Patricia Casas-Agustench
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sara Tulipani
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain.,Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Malaga (IBIMA), Virgen de la Victoria University Hospital,, Málaga University, Malaga 29010, Spain
| | - Antonio Miñarro
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain.,Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, Barcelona 08028, Spain
| | - Alex Sanchez-Pla
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain.,Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, Barcelona 08028, Spain
| | - Mora Murri
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Malaga (IBIMA), Virgen de la Victoria University Hospital,, Málaga University, Malaga 29010, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Francisco J Tinahones
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Malaga (IBIMA), Virgen de la Victoria University Hospital,, Málaga University, Malaga 29010, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
47
|
Coupling Machine Learning and Lipidomics as a Tool to Investigate Metabolic Dysfunction-Associated Fatty Liver Disease. A General Overview. Biomolecules 2021; 11:biom11030473. [PMID: 33810079 PMCID: PMC8004861 DOI: 10.3390/biom11030473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatic biopsy is the gold standard for staging nonalcoholic fatty liver disease (NAFLD). Unfortunately, accessing the liver is invasive, requires a multidisciplinary team and is too expensive to be conducted on large segments of the population. NAFLD starts quietly and can progress until liver damage is irreversible. Given this complex situation, the search for noninvasive alternatives is clinically important. A hallmark of NAFLD progression is the dysregulation in lipid metabolism. In this context, recent advances in the area of machine learning have increased the interest in evaluating whether multi-omics data analysis performed on peripheral blood can enhance human interpretation. In the present review, we show how the use of machine learning can identify sets of lipids as predictive biomarkers of NAFLD progression. This approach could potentially help clinicians to improve the diagnosis accuracy and predict the future risk of the disease. While NAFLD has no effective treatment yet, the key to slowing the progression of the disease may lie in predictive robust biomarkers. Hence, to detect this disease as soon as possible, the use of computational science can help us to make a more accurate and reliable diagnosis. We aimed to provide a general overview for all readers interested in implementing these methods.
Collapse
|
48
|
Bernier-Graveline A, Lesage V, Cabrol J, Lair S, Michaud R, Rosabal M, Verreault J. Lipid metabolites as indicators of body condition in highly contaminant-exposed belugas from the endangered St. Lawrence Estuary population (Canada). ENVIRONMENTAL RESEARCH 2021; 192:110272. [PMID: 33038366 DOI: 10.1016/j.envres.2020.110272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
The endangered St. Lawrence Estuary (SLE) beluga population is declining and has shown no sign of recovery over the past decades despite several protective measures. Changes in the availability of food resources and exposure to organohalogen contaminants have been suggested as potential factors limiting the recovery of this population. Studies on SLE belugas have suggested that contaminant exposure may perturb energy metabolism, however, whether this translates into changes in energy reserves (lipid composition) and body condition is unknown. The objective of this study was to investigate the relationships between body condition and concentrations of organohalogens (polychlorinated biphenyls, organochlorine pesticides, and flame retardants) and a range of lipid metabolites (fatty acids, acylcarnitines, lysophosphatidylcholines, phosphatidylcholines, and sphingomyelins) in blubber samples collected from 51 SLE beluga carcasses recovered between 1998 and 2016 for which the cause of mortality was documented. Blubber Σ9fatty acid concentrations in SLE belugas significantly decreased between 1998 and 2016, suggesting a decline in energy reserves over the past two decades. Concentrations of several phosphatidylcholine analogues were greater in blubber of beluga males and/or females that were in poor body condition compared to those in good body condition. Moreover, concentrations of phosphatidylcholine acyl-alkyl C32:2 were greater in females that died from primary starvation (poor body condition). Greater concentrations of Σ12emerging flame retardants were also found in blubber of SLE beluga females that were in poorer body condition. This study suggests that the use of membrane lipids including phosphatidylcholine concentrations may be a good indicator of body condition and energy reserve status in blubber of marine mammals.
Collapse
Affiliation(s)
- Alexandre Bernier-Graveline
- Centre de recherche en toxicologie de l'environnement (TOXEN), Département des sciences biologiques, Université du Québec à Montréal, P.O. Box 8888, Succursale Centre-ville, Montréal, QC, H3C 3P8, Canada
| | - Véronique Lesage
- Maurice Lamontagne Institute, Fisheries and Oceans Canada, P.O. Box 1000, 850 route de la Mer, Mont-Joli, QC, G5H 3Z4, Canada
| | - Jory Cabrol
- Maurice Lamontagne Institute, Fisheries and Oceans Canada, P.O. Box 1000, 850 route de la Mer, Mont-Joli, QC, G5H 3Z4, Canada
| | - Stéphane Lair
- Canadian Wildlife Health Cooperative, Faculté de médecine vétérinaire, Université de Montréal, St. Hyacinthe, QC, J2S 2M2, Canada
| | - Robert Michaud
- Group for Research and Education on Marine Mammals, Tadoussac, QC, G0T 2A0, Canada
| | - Maikel Rosabal
- Centre de recherche en toxicologie de l'environnement (TOXEN), Département des sciences biologiques, Université du Québec à Montréal, P.O. Box 8888, Succursale Centre-ville, Montréal, QC, H3C 3P8, Canada
| | - Jonathan Verreault
- Centre de recherche en toxicologie de l'environnement (TOXEN), Département des sciences biologiques, Université du Québec à Montréal, P.O. Box 8888, Succursale Centre-ville, Montréal, QC, H3C 3P8, Canada.
| |
Collapse
|
49
|
Li X, Wang X, Li H, Li Y, Guo Y. Seminal Plasma Lipidomics Profiling to Identify Signatures of Kallmann Syndrome. Front Endocrinol (Lausanne) 2021; 12:692690. [PMID: 34393999 PMCID: PMC8358976 DOI: 10.3389/fendo.2021.692690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/27/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Kallmann syndrome (KS) is a rare developmental disorder. Our previous metabolomics work showed substantial changes in linoleic acid and glycerophospholipid metabolism in KS. Here, we performed targeted lipidomics to further identify the differential lipid species in KS. METHODS Twenty-one patients with KS (treatment group) and twenty-two age-matched healthy controls (HC, control group) were enrolled. Seminal plasma samples and medical records were collected. Targeted lipidomics analysis of these samples was performed using ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS). RESULTS Lipidomics profiling of patients with KS and the HCs showed clear separation in the orthogonal projections to latent structures-discriminant analysis (OPLS-DA). There were many differential lipids identified, with the main differential lipid species being triacylglycerols (TAGs), phosphatidylcholines (PCs) and phosphatidylethanolamine (PE). CONCLUSIONS The lipidomics profile of patients with KS changed. It was also determined that TAGs, PCs and PE are promising biomarkers for KS diagnosis. To our knowledge, this is the first report to analyze lipidomics in men with Kallmann syndrome.
Collapse
Affiliation(s)
- Xiaogang Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xi Wang
- National Health Commission (NHC), Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Ye Guo, ; Yongzhe Li,
| | - Ye Guo
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Ye Guo, ; Yongzhe Li,
| |
Collapse
|
50
|
Loizides-Mangold U, Petrenko V, Dibner C. Circadian Lipidomics: Analysis of Lipid Metabolites Around the Clock. Methods Mol Biol 2020; 2130:169-183. [PMID: 33284444 DOI: 10.1007/978-1-0716-0381-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Lipidomics has been defined as the large-scale analysis of lipids in organelles, cells, tissues, or whole organisms. Including the temporal aspects of lipid metabolic changes into this analysis allows to access yet another important aspect of lipid regulation. The resulting methodology, circadian lipidomics, has thus emerged as a novel tool to address the enormous complexity, which is present among cellular lipids. Here, we describe how mass spectrometry-based circadian lipidomics can be applied to study the impact of peripheral clocks on lipid metabolism in human primary cells and tissues, exemplified by studies in human pancreatic islets and skeletal myotubes.
Collapse
Affiliation(s)
- Ursula Loizides-Mangold
- Division of Endocrinology, Diabetes and Nutrition Division, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Volodymyr Petrenko
- Division of Endocrinology, Diabetes and Nutrition Division, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Division of Endocrinology, Diabetes and Nutrition Division, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland.
- Diabetes Center of the Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|