1
|
Holdaway CM, Leonard KA, Nelson R, van der Veen J, Das C, Watts R, Clugston RD, Lehner R, Jacobs RL. Alterations in phosphatidylethanolamine metabolism impacts hepatocellular lipid storage, energy homeostasis, and proliferation. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159608. [PMID: 40154596 DOI: 10.1016/j.bbalip.2025.159608] [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: 11/28/2024] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Phosphatidylethanolamine (PE) is the second most abundant glycerophospholipid in eukaryotic membranes and is involved in several cellular processes. An important pathway for de novo PE synthesis is the Kennedy Pathway. The rate limiting enzyme in the pathway, CTP:phosphoethanolamine cytidyltransferase, catalyzes the synthesis of CDP-ethanolamine from phosphoethanolamine (pEtn) and CTP. Ethanolamine phosphate phospholyase (ETNPPL) has the potential to breakdown pEtn by catabolizing it to form acetaldehyde, ammonium, and inorganic phosphate. Research on this enzyme is limited and it is unclear how its activity affects PE synthesis; therefore, an investigation into ETNPPL's biological effects is required. ETNPPL was expressed in human hepatoma cell line (Huh7) by stable transfection, allowing for long-term expression in cells without ETNPPL. We show that ETNPPL reduces cellular pEtn synthesized from ethanolamine, which decreased synthesis of PE and an increased PC:PE ratio, which has been shown to be associated with metabolic dysfunction-associated steatotic liver disease (MASLD) and impaired mitochondrial function. Experiments conducted show increased neutral lipid storage accompanied by decreased ATP production and oxygen consumption; however, no differences in triglyceride secretion were seen, although ApoB100 secretion was reduced. Huh7 cells expressing ETNPPL proliferate at a slower rate than control and have increased mRNA expression of p53 and tumor suppressor genes (CDKN1A, BBC3, BAX, BRCA1), implicating ETNPPL in cell proliferation, cancer development and/or tumor progression. Overall, ETNPPL rewires hepatic lipid metabolism, altering several processes including increasing lipid storage and decreasing proliferation. The impacts observed in this study may create a link between hepatic ETNPPL expression and MASLD/HCC pathophysiology.
Collapse
Affiliation(s)
- Courtney M Holdaway
- Department of Agricultural, Food & Nutritional Science, 4-002 Li Ka Shing Center for Health and Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Kelly-Ann Leonard
- Department of Agricultural, Food & Nutritional Science, 4-002 Li Ka Shing Center for Health and Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Randal Nelson
- Department of Pediatrics, 315 Heritage Medical Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Jelske van der Veen
- Department of Agricultural, Food & Nutritional Science, 4-002 Li Ka Shing Center for Health and Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Chinmayee Das
- Department of Pediatrics, 315 Heritage Medical Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Russell Watts
- Department of Pediatrics, 315 Heritage Medical Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Robin D Clugston
- Department of Physiology, 7-49 Medical Sciences Building, University of Alberta, Edmonton, AB T6G 2H7, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Richard Lehner
- Department of Pediatrics, 315 Heritage Medical Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada; Group on the Molecular and Cell Biology of Lipids, Canada
| | - Rene L Jacobs
- Department of Agricultural, Food & Nutritional Science, 4-002 Li Ka Shing Center for Health and Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada; Group on the Molecular and Cell Biology of Lipids, Canada.
| |
Collapse
|
2
|
Liu L, Cui H, Dong N, Zhu X, Li S, Ma X, Niu D. Effects of phosphatidylethanolamine on intramuscular fat deposition and key gene identification by transcriptome sequencing in broiler chickens. Poult Sci 2025; 104:104914. [PMID: 39983529 PMCID: PMC11889968 DOI: 10.1016/j.psj.2025.104914] [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: 11/29/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025] Open
Abstract
High-intensity breeding of broilers has increased meat yield while reducing meat quality. Properly enhancing intramuscular fat (IMF) content is an effective strategy to improve chicken meat quality and boost consumer preference. Given that the role of phosphatidylethanolamine (PE) in chicken IMF metabolism remains unclear, we investigated the effects of PE on the meat quality, as well as the differentiation and gene expression regulation of chicken intramuscular adipocytes (IMAs). PE supplementation significantly increased the a* value of meat color and reduced shear force (P < 0.05); however, it did not exert a significant effect on the pH value 45 min post-slaughter (P > 0.05). After treating intramuscular adipocytes IMAs with 25, 50, and 100 µM PE, 100 µM PE supplement markedly enhanced lipid deposition and the expression of genes related to adipogenic differentiation. Differentially expressed genes (DEGs) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GESA) were used to identify key genes involved in PE regulation of chicken IMA deposition. A total of 1,182 DEGs were identified, with genes S1PR3, FABP4, PLIN2, APOA1, PPARG and CD36 recognized as hub genes associated with the triglycerides (TG) content of IMAs. PPAR signaling pathway, terpenoid backbone biosynthesis, cytokine-cytokine receptor interaction, and neuroactive ligand-receptor interaction pathway were significantly enriched in PE group. This study reveals the role of PE in chicken IMAs differentiation and lipid deposition, providing a theoretical foundation for further research into the impact of PE on IMF accumulation in broiler chickens.
Collapse
Affiliation(s)
- Lu Liu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Houxue Cui
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Nanxi Dong
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Xianghua Zhu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Sihong Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China.
| |
Collapse
|
3
|
Hussain M, Khan I, Chaudhary MN, Ali K, Mushtaq A, Jiang B, Zheng L, Pan Y, Hu J, Zou X. Phosphatidylserine: A comprehensive overview of synthesis, metabolism, and nutrition. Chem Phys Lipids 2024; 264:105422. [PMID: 39097133 DOI: 10.1016/j.chemphyslip.2024.105422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Phosphatidylserine (PtdS) is classified as a glycerophospholipid and a primary anionic phospholipid and is particularly abundant in the inner leaflet of the plasma membrane in neural tissues. It is synthesized from phosphatidylcholine or phosphatidylethanolamine by exchanging the base head group with serine, and this reaction is catalyzed by PtdS synthase-1 and PtdS synthase-2 located in the endoplasmic reticulum. PtdS exposure on the outside surface of the cell is essential for eliminating apoptotic cells and initiating the blood clotting cascade. It is also a precursor of phosphatidylethanolamine, produced by PtdS decarboxylase in bacteria, yeast, and mammalian cells. Furthermore, PtdS acts as a cofactor for several necessary enzymes that participate in signaling pathways. Beyond these functions, several studies indicate that PtdS plays a role in various cerebral functions, including activating membrane signaling pathways, neuroinflammation, neurotransmission, and synaptic refinement associated with the central nervous system (CNS). This review discusses the occurrence of PtdS in nature and biosynthesis via enzymes and genes in plants, yeast, prokaryotes, mammalian cells, and the brain, and enzymatic synthesis through phospholipase D (PLD). Furthermore, we discuss metabolism, its role in the CNS, the fortification of foods, and supplementation for improving some memory functions, the results of which remain unclear. PtdS can be a potentially beneficial addition to foods for kids, seniors, athletes, and others, especially with the rising consumer trend favoring functional foods over conventional pills and capsules. Clinical studies have shown that PtdS is safe and well tolerated by patients.
Collapse
Affiliation(s)
- Mudassar Hussain
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Imad Khan
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Muneeba Naseer Chaudhary
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing, 400715, China
| | - Khubaib Ali
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Anam Mushtaq
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Bangzhi Jiang
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Lei Zheng
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yuechao Pan
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jijie Hu
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xiaoqiang Zou
- State Key Laboratory of Food Science and Resources, National Engineering Research Center for Functional Food, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
4
|
Yan Z, Zhang Y, Nan N, Ji S, Lan S, Qin G, Sang N. YTHDC2 mediated RNA m 6A modification contributes to PM 2.5-induced hepatic steatosis. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135004. [PMID: 38943883 DOI: 10.1016/j.jhazmat.2024.135004] [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: 04/01/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
Exposure to fine particulate matter (PM2.5) is a significant risk factor for hepatic steatosis. The N6-methyladenosine (m6A) is implicated in metabolic disturbances triggered by exogenous environmental factors. However, the role of m6A in mediating PM2.5-induced hepatic steatosis remains unclear. Herein, male C57BL/6J mice were subjected to PM2.5 exposure throughout the entire heating season utilizing a real-ambient PM2.5 whole-body inhalation exposure system. Concurrently, HepG2 cell models exposed to PM2.5 were developed to delve the role of m6A methylation modification. Following PM2.5 exposure, significant hepatic lipid accumulation and elevated global m6A level were observed both in vitro and in vivo. The downregulation of YTHDC2, an m6A-binding protein, might contribute to this alteration. In vitro studies revealed that lipid-related genes CEPT1 and YWHAH might be targeted by m6A modification. YTHDC2 could bind to CDS region of them and increase their stability. Exposure to PM2.5 shortened mRNA lifespan and suppressed the expression of CEPT1 and YWHAH, which were reversed to baseline or higher level upon the enforced expression of YTHDC2. Consequently, our findings indicate that PM2.5 induces elevated m6A methylation modification of CEPT1 and YWHAH by downregulating YTHDC2, which in turn mediates the decrease in the mRNA stabilization and expression of these genes, ultimately resulting in hepatic steatosis.
Collapse
Affiliation(s)
- Zhipeng Yan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Yaru Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Nan Nan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Shaoyang Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Siyi Lan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| |
Collapse
|
5
|
Duncan RE. Deficiency of phosphatidylethanolamine synthesis: consequences for skeletal muscle. FUNCTION 2023; 4:zqad044. [PMID: 37772311 PMCID: PMC10533200 DOI: 10.1093/function/zqad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/30/2023] Open
Affiliation(s)
- Robin Elaine Duncan
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
6
|
Anari M, Montgomery MK. Phospholipid metabolism in the liver - Implications for phosphatidylserine in non-alcoholic fatty liver disease. Biochem Pharmacol 2023; 213:115621. [PMID: 37217141 DOI: 10.1016/j.bcp.2023.115621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Mammalian cells contain more than a thousand different glycerophospholipid species that are essential membrane components and signalling molecules, with phosphatidylserine (PS) giving membranes their negative surface charge. Depending on the tissue, PS is important in apoptosis, blood clotting, cancer pathogenesis, as well as muscle and brain function, processes that are dependent on the asymmetrical distribution of PS on the plasma membrane and/or the capacity of PS to act as anchorage for various signalling proteins. Recent studies have implicated hepatic PS in the progression of non-alcoholic fatty liver disease (NAFLD), either as beneficial in the context of suppressing hepatic steatosis and fibrosis, or on the other hand as a potential contributor to the progression of liver cancer. This review provides an extensive overview of hepatic phospholipid metabolism, including its biosynthetic pathways, intracellular trafficking and roles in health and disease, further taking a deeper dive into PS metabolism, including associate and causative evidence of the role of PS in advanced liver disease.
Collapse
Affiliation(s)
- Marziyeh Anari
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
7
|
Papadopoulos G, Legaki AI, Georgila K, Vorkas P, Giannousi E, Stamatakis G, Moustakas II, Petrocheilou M, Pyrina I, Gercken B, Kassi E, Chavakis T, Pateras IS, Panayotou G, Gika H, Samiotaki M, Eliopoulos AG, Chatzigeorgiou A. Integrated omics analysis for characterization of the contribution of high fructose corn syrup to non-alcoholic fatty liver disease in obesity. Metabolism 2023; 144:155552. [PMID: 36996933 DOI: 10.1016/j.metabol.2023.155552] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND High-Fructose Corn Syrup (HFCS), a sweetener rich in glucose and fructose, is nowadays widely used in beverages and processed foods; its consumption has been correlated to the emergence and progression of Non-Alcoholic Fatty Liver Disease (NAFLD). Nevertheless, the molecular mechanisms by which HFCS impacts hepatic metabolism remain scarce, especially in the context of obesity. Besides, the majority of current studies focuses either on the detrimental role of fructose in hepatic steatosis or compare separately the additive impact of fructose versus glucose in high fat diet-induced NAFLD. AIM By engaging combined omics approaches, we sought to characterize the role of HFCS in obesity-associated NAFLD and reveal molecular processes, which mediate the exaggeration of steatosis under these conditions. METHODS Herein, C57BL/6 mice were fed a normal-fat-diet (ND), a high-fat-diet (HFD) or a HFD supplemented with HFCS (HFD-HFCS) and upon examination of their metabolic and NAFLD phenotype, proteomic, lipidomic and metabolomic analyses were conducted to identify HFCS-related molecular alterations of the hepatic metabolic landscape in obesity. RESULTS Although HFD and HFD-HFCS mice displayed comparable obesity, HFD-HFCS mice showed aggravation of hepatic steatosis, as analysis of the lipid droplet area in liver sections revealed (12,15 % of total section area in HFD vs 22,35 % in HFD-HFCS), increased NAFLD activity score (3,29 in HFD vs 4,86 in HFD-HFCS) and deteriorated hepatic insulin resistance, as compared to the HFD mice. Besides, the hepatic proteome of HFD-HFCS mice was characterized by a marked upregulation of 5 core proteins implicated in de novo lipogenesis (DNL), while an increased phosphatidyl-cholines(PC)/phosphatidyl-ethanolamines(PE) ratio (2.01 in HFD vs 3.04 in HFD-HFCS) was observed in the livers of HFD-HFCS versus HFD mice. Integrated analysis of the omics datasets indicated that Tricarboxylic Acid (TCA) cycle overactivation is likely contributing towards the intensification of steatosis during HFD-HFCS-induced NAFLD. CONCLUSION Our results imply that HFCS significantly contributes to steatosis aggravation during obesity-related NAFLD, likely deriving from DNL upregulation, accompanied by TCA cycle overactivation and deteriorated hepatic insulin resistance.
Collapse
Affiliation(s)
- Grigorios Papadopoulos
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Aigli-Ioanna Legaki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Konstantina Georgila
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Vorkas
- Institute of Applied Biosciences, Centre for Research and Technology, 57001, Thermi, Thessaloniki, Greece
| | - Eirini Giannousi
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - George Stamatakis
- Institute for Bio-innovation, Biomedical Sciences Research Center "Alexander Fleming", Vari 16672, Greece
| | - Ioannis I Moustakas
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Maria Petrocheilou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001, Thermi, Thessaloniki, Greece
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Ioannis S Pateras
- 2nd Department of Pathology, "Attikon" University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - George Panayotou
- Institute for Bio-innovation, Biomedical Sciences Research Center "Alexander Fleming", Vari 16672, Greece
| | - Helen Gika
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001, Thermi, Thessaloniki, Greece
| | - Martina Samiotaki
- Institute for Bio-innovation, Biomedical Sciences Research Center "Alexander Fleming", Vari 16672, Greece
| | - Aristides G Eliopoulos
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece; Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
8
|
Basu S, Pawlowic MC, Hsu FF, Thomas G, Zhang K. Ethanolaminephosphate cytidylyltransferase is essential for survival, lipid homeostasis and stress tolerance in Leishmania major. PLoS Pathog 2023; 19:e1011112. [PMID: 37506172 PMCID: PMC10411802 DOI: 10.1371/journal.ppat.1011112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Glycerophospholipids including phosphatidylethanolamine (PE) and phosphatidylcholine (PC) are vital components of biological membranes. Trypanosomatid parasites of the genus Leishmania can acquire PE and PC via de novo synthesis and the uptake/remodeling of host lipids. In this study, we investigated the ethanolaminephosphate cytidylyltransferase (EPCT) in Leishmania major, which is the causative agent for cutaneous leishmaniasis. EPCT is a key enzyme in the ethanolamine branch of the Kennedy pathway which is responsible for the de novo synthesis of PE. Our results demonstrate that L. major EPCT is a cytosolic protein capable of catalyzing the formation of CDP-ethanolamine from ethanolamine-phosphate and cytidine triphosphate. Genetic manipulation experiments indicate that EPCT is essential in both the promastigote and amastigote stages of L. major as the chromosomal null mutants cannot survive without the episomal expression of EPCT. This differs from our previous findings on the choline branch of the Kennedy pathway (responsible for PC synthesis) which is required only in promastigotes but not amastigotes. While episomal EPCT expression does not affect promastigote proliferation under normal conditions, it leads to reduced production of ethanolamine plasmalogen or plasmenylethanolamine, the dominant PE subtype in Leishmania. In addition, parasites with episomal EPCT exhibit heightened sensitivity to acidic pH and starvation stress, and significant reduction in virulence. In summary, our investigation demonstrates that proper regulation of EPCT expression is crucial for PE synthesis, stress response, and survival of Leishmania parasites throughout their life cycle.
Collapse
Affiliation(s)
- Somrita Basu
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Mattie C. Pawlowic
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America
- Wellcome Centre for Anti-Infectives Research (WCAIR), Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Geoff Thomas
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America
| |
Collapse
|
9
|
Li L, Song Y, Shi Y, Sun L. Thyroid Hormone Receptor-β Agonists in NAFLD Therapy: Possibilities and Challenges. J Clin Endocrinol Metab 2023; 108:1602-1613. [PMID: 36746649 DOI: 10.1210/clinem/dgad072] [Citation(s) in RCA: 18] [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: 09/04/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive metabolic liver disease with an unknown pathogenesis and no FDA-approved drug treatment to date. Hypothyroidism has been identified as a risk factor for NAFLD as thyroxine is required for regulating metabolism in adults. Thyroxine has been shown to reduce fat in the livers of murine models with experimentally induced NAFLD. The use of synthetic thyroxine has been shown to increase lipid metabolism leading to weight loss; however, thyroxine has also been shown to cause many side effects, especially in the heart. Overcoming these cardiac side effects involves designing agonists specific to one of the 2 gene subtypes for the thyroid hormone (TH) receptor (TR), TRβ. While the other TH receptor subtype, TRα, is mainly expressed in the heart and is responsible for thyroxine's cardiac function, TRβ is mainly expressed in the liver and is involved in liver function. Using TRβ-specific agonists to treat NAFLD can prevent cardiac and other adverse side effects. Several TRβ-specific agonists have shown positive therapeutic effects in NAFLD animal models and have entered clinical trials. We seek to provide a comprehensive updated reference of TRβ-specific agonists in this review and explore the future therapeutic potential of TRβ-specific activation in the treatment of NAFLD.
Collapse
Affiliation(s)
- Limei Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Yan Song
- Department of Endocrinology and Metabolism, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yongquan Shi
- Department of Endocrinology and Metabolism, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Liangliang Sun
- Department of Endocrinology and Metabolism, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
10
|
Grapentine S, Singh RK, Bakovic M. Skeletal Muscle Consequences of Phosphatidylethanolamine Synthesis Deficiency. FUNCTION 2023; 4:zqad020. [PMID: 37342414 PMCID: PMC10278983 DOI: 10.1093/function/zqad020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 06/22/2023] Open
Abstract
The maintenance of phospholipid homeostasis is increasingly being implicated in metabolic health. Phosphatidylethanolamine (PE) is the most abundant phospholipid on the inner leaflet of cellular membranes, and we have previously shown that mice with a heterozygous ablation of the PE synthesizing enzyme, Pcyt2 (Pcyt2+/-), develop obesity, insulin resistance, and NASH. Skeletal muscle is a major determinant of systemic energy metabolism, making it a key player in metabolic disease development. Both the total PE levels and the ratio of PE to other membrane lipids in skeletal muscle are implicated in insulin resistance; however, the underlying mechanisms and the role of Pcyt2 regulation in this association remain unclear. Here, we show how reduced phospholipid synthesis due to Pcyt2 deficiency causes Pcyt2+/- skeletal muscle dysfunction and metabolic abnormalities. Pcyt2+/- skeletal muscle exhibits damage and degeneration, with skeletal muscle cell vacuolization, disordered sarcomeres, mitochondria ultrastructure irregularities and paucity, inflammation, and fibrosis. There is intramuscular adipose tissue accumulation, and major disturbances in lipid metabolism with impaired FA mobilization and oxidation, elevated lipogenesis, and long-chain fatty acyl-CoA, diacylglycerol, and triacylglycerol accumulation. Pcyt2+/- skeletal muscle exhibits perturbed glucose metabolism with elevated glycogen content, impaired insulin signaling, and reduced glucose uptake. Together, this study lends insight into the critical role of PE homeostasis in skeletal muscle metabolism and health with broad implications on metabolic disease development.
Collapse
Affiliation(s)
- Sophie Grapentine
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Rathnesh K Singh
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| |
Collapse
|
11
|
Cikes D, Elsayad K, Sezgin E, Koitai E, Torma F, Orthofer M, Yarwood R, Heinz LX, Sedlyarov V, Miranda ND, Taylor A, Grapentine S, Al-Murshedi F, Abot A, Weidinger A, Kutchukian C, Sanchez C, Cronin SJF, Novatchkova M, Kavirayani A, Schuetz T, Haubner B, Haas L, Hagelkruys A, Jackowski S, Kozlov AV, Jacquemond V, Knauf C, Superti-Furga G, Rullman E, Gustafsson T, McDermot J, Lowe M, Radak Z, Chamberlain JS, Bakovic M, Banka S, Penninger JM. PCYT2-regulated lipid biosynthesis is critical to muscle health and ageing. Nat Metab 2023; 5:495-515. [PMID: 36941451 DOI: 10.1038/s42255-023-00766-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2023] [Indexed: 03/23/2023]
Abstract
Muscle degeneration is the most prevalent cause for frailty and dependency in inherited diseases and ageing. Elucidation of pathophysiological mechanisms, as well as effective treatments for muscle diseases, represents an important goal in improving human health. Here, we show that the lipid synthesis enzyme phosphatidylethanolamine cytidyltransferase (PCYT2/ECT) is critical to muscle health. Human deficiency in PCYT2 causes a severe disease with failure to thrive and progressive weakness. pcyt2-mutant zebrafish and muscle-specific Pcyt2-knockout mice recapitulate the participant phenotypes, with failure to thrive, progressive muscle weakness and accelerated ageing. Mechanistically, muscle Pcyt2 deficiency affects cellular bioenergetics and membrane lipid bilayer structure and stability. PCYT2 activity declines in ageing muscles of mice and humans, and adeno-associated virus-based delivery of PCYT2 ameliorates muscle weakness in Pcyt2-knockout and old mice, offering a therapy for individuals with a rare disease and muscle ageing. Thus, PCYT2 plays a fundamental and conserved role in vertebrate muscle health, linking PCYT2 and PCYT2-synthesized lipids to severe muscle dystrophy and ageing.
Collapse
Affiliation(s)
- Domagoj Cikes
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.
| | - Kareem Elsayad
- Division of Anatomy, Center for Anatomy and Cell Biology and Medical Imaging Cluster (MIC), Vienna, Austria.
| | - Erdinc Sezgin
- MRC Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, Oxford, UK
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Erika Koitai
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Ferenc Torma
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Michael Orthofer
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Rebecca Yarwood
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Leonhard X Heinz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Adrian Taylor
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Sophie Grapentine
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Fathiya Al-Murshedi
- Department of Genetics, College of Medicine, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Anne Abot
- Enterosys SAS, Prologue Biotech, Labège, France
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Candice Kutchukian
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Colline Sanchez
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Shane J F Cronin
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Maria Novatchkova
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Anoop Kavirayani
- VBCF, Vienna BioCenter Core Facilities, Vienna BioCenter, Vienna, Austria
| | - Thomas Schuetz
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Bernhard Haubner
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisa Haas
- IMP Research Institute of Molecular Pathology, Vienna, Austria
| | - Astrid Hagelkruys
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Vincent Jacquemond
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Claude Knauf
- INSERM U1220 Institut de Recherche en Santé Digestive, CHU Purpan, Université Toulouse III Paul Sabatier Toulouse, Toulouse, France
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Eric Rullman
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
- Cardiovascular Theme, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Thomas Gustafsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - John McDermot
- Manchester Centre for Genomics Medicine, St Mary's Hospital, Manchester University Hospital Foundation Trust, Manchester, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington, Seattle, WA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, WA, USA
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Siddharth Banka
- Manchester Centre for Genomics Medicine, St Mary's Hospital, Manchester University Hospital Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.
- Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Chen Y, Jiang H, Zhan Z, Lu J, Gu T, Yu P, Liang W, Zhang X, Liu S, Bi H, Zhong S, Tang L. Restoration of lipid homeostasis between TG and PE by the LXRα-ATGL/EPT1 axis ameliorates hepatosteatosis. Cell Death Dis 2023; 14:85. [PMID: 36746922 PMCID: PMC9902534 DOI: 10.1038/s41419-023-05613-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/08/2023]
Abstract
Converting lipid disturbances in response to energy oversupply into healthy lipid homeostasis is a promising therapy to alleviate hepatosteatosis. Our clinical studies found that a further elevation of triglyceride (TG) in obese patients with the body mass index (BMI) greater than 28 was accompanied by a further reduction of phosphatidylethanolamine (PE). Shorter survival and poor prognosis were shown for the patients with high TG and low PE levels. Liver X receptor alpha (LXRα) knockout mice aggravated high-fat diet (HFD)-induced obesity and lipid disorders, making the TG enrichment and the PE decrease more pronounced according to the liver lipidomics analysis. The RNA-seq from mice liver exhibited that these metabolism disorders were attributed to the decline of Atgl (encoding the TG metabolism enzyme ATGL) and Ept1 (encoding the PE synthesis enzyme EPT1) expression. Mechanistic studies uncovered that LXRα activated the ATGL and EPT1 gene via direct binding to a LXR response element (LXRE) in the promoter. Moreover, both the supplement of PE in statin or fibrate therapy, and the LXRα inducer (oridonin) ameliorated cellular lipid deposition and lipotoxicity. Altogether, restoration of lipid homeostasis of TG and PE via the LXRα-ATGL/EPT1 axis may be a potential approach for the management of hepatosteatosis and metabolic syndrome.
Collapse
Affiliation(s)
- Yulian Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Huanguo Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Zhikun Zhan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Jindi Lu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Tanwei Gu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Ping Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Weimin Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Xi Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Shuwen Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Shilong Zhong
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China.
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Lan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
13
|
Xu H, Li W, Huang L, He X, Xu B, He X, Chen W, Wang Y, Xu W, Wang S, Kong Q, Xu Y, Lu W. Phosphoethanolamine cytidylyltransferase ameliorates mitochondrial function and apoptosis in hepatocytes in T2DM in vitro. J Lipid Res 2023; 64:100337. [PMID: 36716821 PMCID: PMC10033998 DOI: 10.1016/j.jlr.2023.100337] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/29/2023] Open
Abstract
Liver function indicators are often impaired in patients with type 2 diabetes mellitus (T2DM), who present higher concentrations of aspartate aminotransferase, alanine aminotransferase, and gamma-glutamyl transferase than individuals without diabetes. However, the mechanism of liver injury in patients with T2DM has not been clearly elucidated. In this study, we performed a lipidomics analysis on the liver of T2DM mice, and we found that phosphatidylethanolamine (PE) levels were low in T2DM, along with an increase in diglyceride, which may be due to a decrease in the levels of phosphoethanolamine cytidylyltransferase (Pcyt2), thus likely affecting the de novo synthesis of PE. The phosphatidylserine decarboxylase pathway did not change significantly in the T2DM model, although both pathways are critical sources of PE. Supplementation with CDP-ethanolamine (CDP-etn) to increase the production of PE from the CDP-etn pathway reversed high glucose and FFA (HG&FFA)-induced mitochondrial damage including increased apoptosis, decreased ATP synthesis, decreased mitochondrial membrane potential, and increased reactive oxygen species, whereas supplementation with lysophosphatidylethanolamine, which can increase PE production in the phosphatidylserine decarboxylase pathway, did not. Additionally, we found that overexpression of PCYT2 significantly ameliorated ATP synthesis and abnormal mitochondrial morphology induced by HG&FFA. Finally, the BAX/Bcl-2/caspase3 apoptosis pathway was activated in hepatocytes of the T2DM model, which could also be reversed by CDP-etn supplements and PCYT2 overexpression. In summary, in the liver of T2DM mice, Pcyt2 reduction may lead to a decrease in the levels of PE, whereas CDP-etn supplementation and PCYT2 overexpression ameliorate partial mitochondrial function and apoptosis in HG&FFA-stimulated L02 cells.
Collapse
Affiliation(s)
- Hu Xu
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Weizu Li
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Lei Huang
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Xinyu He
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Bei Xu
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Xueqing He
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Wentong Chen
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Yaoxing Wang
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Wenjun Xu
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Sheng Wang
- Center for Scientific Research, Anhui Medical University, Hefei, China
| | - Qin Kong
- Basic Medical College, Anhui Medical University, Hefei, China
| | - Youzhi Xu
- Basic Medical College, Anhui Medical University, Hefei, China.
| | - Wenjie Lu
- Basic Medical College, Anhui Medical University, Hefei, China.
| |
Collapse
|
14
|
Basu S, Pawlowic M, Hsu FF, Thomas G, Zhang K. Ethanolaminephosphate cytidyltransferase is essential for survival, lipid homeostasis and stress tolerance in Leishmania major. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523530. [PMID: 36712124 PMCID: PMC9882048 DOI: 10.1101/2023.01.10.523530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Glycerophospholipids including phosphatidylethanolamine (PE) and phosphatidylcholine (PC) are vital components of biological membranes. Trypanosomatid parasites of the genus Leishmania can acquire PE and PC via de novo synthesis and the uptake/remodeling of host lipids. In this study, we investigated the ethanolaminephosphate cytidyltransferase (EPCT) in Leishmania major , which is the causative agent for cutaneous leishmaniasis. EPCT is a key enzyme in the ethanolamine branch of the Kennedy pathway which is responsible for the de novo synthesis of PE. Our results demonstrate that L. major EPCT is a cytosolic protein capable of catalyzing the formation of CDP-ethanolamine from ethanolamine-phosphate and cytidine triphosphate. Genetic manipulation experiments indicate that EPCT is essential in both the promastigote and amastigote stages of L. major as the chromosomal null mutants cannot survive without the episomal expression of EPCT. This differs from our previous findings on the choline branch of the Kennedy pathway (responsible for PC synthesis) which is required only in promastigotes but not amastigotes. While episomal EPCT expression does not affect promastigote proliferation under normal conditions, it leads to reduced production of ethanolamine plasmalogen or plasmenylethanolamine, the dominant PE subtype in Leishmania . In addition, parasites with epsiomal EPCT exhibit heightened sensitivity to acidic pH and starvation stress, and significant reduction in virulence. In summary, our investigation demonstrates that proper regulation of EPCT expression is crucial for PE synthesis, stress response, and survival of Leishmania parasites throughout their life cycle. AUTHOR SUMMARY In nature, Leishmania parasites alternate between fast replicating, extracellular promastigotes in sand fly gut and slow growing, intracellular amastigotes in macrophages. Previous studies suggest that promastigotes acquire most of their lipids via de novo synthesis whereas amastigotes rely on the uptake and remodeling of host lipids. Here we investigated the function of ethanolaminephosphate cytidyltransferase (EPCT) which catalyzes a key step in the de novo synthesis of phosphatidylethanolamine (PE) in Leishmania major . Results showed that EPCT is indispensable for both promastigotes and amastigotes, indicating that de novo PE synthesis is still needed at certain capacity for the intracellular form of Leishmania parasites. In addition, elevated EPCT expression alters overall PE synthesis and compromises parasite’s tolerance to adverse conditions and is deleterious to the growth of intracellular amastigotes. These findings provide new insight into how Leishmania acquire essential phospholipids and how disturbance of lipid metabolism can impact parasite fitness.
Collapse
Affiliation(s)
- Somrita Basu
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Mattie Pawlowic
- Wellcome Centre for Anti-Infectives Research (WCAIR), Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Diabetes, Metabolism, and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, 660S. Euclid Ave., Saint Louis, MO 63110, USA
| | - Geoff Thomas
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
15
|
St Germain M, Iraji R, Bakovic M. Phosphatidylethanolamine homeostasis under conditions of impaired CDP-ethanolamine pathway or phosphatidylserine decarboxylation. Front Nutr 2023; 9:1094273. [PMID: 36687696 PMCID: PMC9849821 DOI: 10.3389/fnut.2022.1094273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/14/2022] [Indexed: 01/07/2023] Open
Abstract
Phosphatidylethanolamine is the major inner-membrane lipid in the plasma and mitochondrial membranes. It is synthesized in the endoplasmic reticulum from ethanolamine and diacylglycerol (DAG) by the CDP-ethanolamine pathway and from phosphatidylserine by decarboxylation in the mitochondria. Recently, multiple genetic disorders that impact these pathways have been identified, including hereditary spastic paraplegia 81 and 82, Liberfarb syndrome, and a new type of childhood-onset neurodegeneration-CONATOC. Individuals with these diseases suffer from multisystem disorders mainly affecting neuronal function. This indicates the importance of maintaining proper phospholipid homeostasis when major biosynthetic pathways are impaired. This study summarizes the current knowledge of phosphatidylethanolamine metabolism in order to identify areas of future research that might lead to the development of treatment options.
Collapse
|
16
|
Fang W, Liu Y, Chen Q, Xu D, Liu Q, Cao X, Hao T, Zhang L, Mai K, Ai Q. Palmitic acid induces intestinal lipid metabolism disorder, endoplasmic reticulum stress and inflammation by affecting phosphatidylethanolamine content in large yellow croaker Larimichthys crocea. Front Immunol 2022; 13:984508. [PMID: 36059525 PMCID: PMC9437641 DOI: 10.3389/fimmu.2022.984508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022] Open
Abstract
In the 21st century, intestinal homeostatic imbalance has emerged as a growing health challenge worldwide. Accumulating evidence reveals that excessive intake of saturated fatty acid (SFA) induces intestinal homeostatic imbalance. However, the potential molecular mechanism is still unclear. In the present study, we found that palm oil or palmitic acid (PA) treatment disturbed lipid metabolism homeostasis and triggered endoplasmic reticulum (ER) stress and inflammation in the intestine or intestinal cells of large yellow croaker (Larimichthys crocea). Interestingly, PA treatment significantly decreased phosphatidylethanolamine (PE) content in the intestinal cells. PE supplementation decreased triglyceride content in the intestinal cells induced by PA treatment by inhibiting fatty acid uptake and lipogenesis. PE supplementation suppressed ER stress. Meanwhile, PE supplementation alleviated inflammatory response through p38 MAPK-p65 pathway, reducing the damage of intestinal cells caused by PA treatment to some extent. Our work revealed that intestinal homeostatic imbalance caused by PA treatment was partly due to the decrease of PE content. PE consumption might be a nutritional strategy to regulate intestinal homeostasis in fish and even human beings.
Collapse
Affiliation(s)
- Wei Fang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Yongtao Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Qiuchi Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Dan Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Xiufei Cao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Tingting Hao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Lu Zhang
- Tongwei Co., Ltd., Chengdu, China
- Healthy Aquaculture Key Laboratory of Sichuan Province, Chengdu, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Qinghui Ai,
| |
Collapse
|
17
|
Slightly different metabolomic profiles are associated with high or low weight duck foie gras. PLoS One 2022; 17:e0255707. [PMID: 35763459 PMCID: PMC9239462 DOI: 10.1371/journal.pone.0255707] [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: 07/20/2021] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
Understanding the evolution of fatty liver metabolism of ducks is a recurrent issue for researchers and industry. Indeed, the increase in weight during the overfeeding period leads to an important change in the liver metabolism. However, liver weight is highly variable at the end of overfeeding within a batch of animals reared, force-fed and slaughtered in the same way. For this study, we performed a proton nuclear magnetic resonance (1H-NMR) analysis on two classes of fatty liver samples, called low-weight liver (weights between 550 and 599 g) and high-weight liver (weights above 700 g). The aim of this study was to identify the differences in metabolism between two classes of liver weight (low and high). Firstly, the results suggested that increased liver weight is associated with higher glucose uptake leading to greater lipid synthesis. Secondly, this increase is probably also due to a decline in the level of export of triglycerides from the liver by maintaining them at high hepatic concentration levels, but also of hepatic cholesterol. Finally, the increase in liver weight could lead to a significant decrease in the efficiency of aerobic energy metabolism associated with a significant increase in the level of oxidative stress. However, all these hypotheses will have to be confirmed in the future, by studies on plasma levels and specific assays to validate these results.
Collapse
|
18
|
Bauer KC, Littlejohn PT, Ayala V, Creus-Cuadros A, Finlay BB. Nonalcoholic Fatty Liver Disease and the Gut-Liver Axis: Exploring an Undernutrition Perspective. Gastroenterology 2022; 162:1858-1875.e2. [PMID: 35248539 DOI: 10.1053/j.gastro.2022.01.058] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic condition affecting one quarter of the global population. Although primarily linked to obesity and metabolic syndrome, undernutrition and the altered (dysbiotic) gut microbiome influence NAFLD progression. Both undernutrition and NAFLD prevalence are predicted to considerably increase, but how the undernourished gut microbiome contributes to hepatic pathophysiology remains far less studied. Here, we present undernutrition conditions with fatty liver features, including kwashiorkor and micronutrient deficiency. We then review the gut microbiota-liver axis, highlighting key pathways linked to NAFLD progression within both overnutrition and undernutrition. To conclude, we identify challenges and collaborative possibilities of emerging multiomic research addressing the pathology and treatment of undernourished NAFLD.
Collapse
Affiliation(s)
- Kylynda C Bauer
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Thoracic and Gastrointestinal Malignancies Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
| | - Paula T Littlejohn
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain; Department of Experimental Medicine, Universitat de Lleida, Lleida, Spain
| | - Anna Creus-Cuadros
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
19
|
Characterization and Roles of Membrane Lipids in Fatty Liver Disease. MEMBRANES 2022; 12:membranes12040410. [PMID: 35448380 PMCID: PMC9025760 DOI: 10.3390/membranes12040410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022]
Abstract
Obesity has reached global epidemic proportions and it affects the development of insulin resistance, type 2 diabetes, fatty liver disease and other metabolic diseases. Membrane lipids are important structural and signaling components of the cell membrane. Recent studies highlight their importance in lipid homeostasis and are implicated in the pathogenesis of fatty liver disease. Here, we discuss the numerous membrane lipid species and their metabolites including, phospholipids, sphingolipids and cholesterol, and how dysregulation of their composition and physiology contribute to the development of fatty liver disease. The development of new genetic and pharmacological mouse models has shed light on the role of lipid species on various mechanisms/pathways; these lipids impact many aspects of the pathophysiology of fatty liver disease and could potentially be targeted for the treatment of fatty liver disease.
Collapse
|
20
|
Huang NK, Matthan NR, Matuszek G, Lichtenstein AH. Plasma Metabolite Profiles Following Consumption of Animal Protein and Soybean-Based Diet in Hypercholesterolemic Postmenopausal Women. Metabolites 2022; 12:209. [PMID: 35323651 PMCID: PMC8952012 DOI: 10.3390/metabo12030209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 02/05/2023] Open
Abstract
Subjective reporting of food intake can be unreliable. No objective method is available to distinguish between diets differing in protein type. To address this gap, a secondary analysis of a randomized controlled cross-over feeding trial was conducted. Assessed were fasting plasma metabolite profiles and their associations with cardiometabolic risk factors (CMRFs). Hypercholesterolemic post-menopausal women (N = 11) were provided with diets containing predominantly animal protein (AP) and soy protein (SP). Untargeted metabolomics were used to determine the plasma metabolite profiles at the end of each diet phase. Concentrations of identified metabolites (N = 829) were compared using paired t-tests adjusted for false discovery rate, partial least square-discrimination analysis (PLS-DA) and receiver operating characteristics (ROC). Among the identified metabolites, 58 differed significantly between the AP and SP diets; the majority were phospholipids (n = 36), then amino acids (n = 10), xenobiotics (n = 7), vitamin/vitamin-related (n = 3) and lipids (n = 2). Of the top 10 metabolites, amino acid-derived metabolites, phospholipids and xenobiotics comprised the main categories differing due to dietary protein type. ROC curves confirmed that the top 10 metabolites were potential discriminating biomarkers for AP- and SP-rich diets. In conclusion, amino acid-derived metabolites, phosphatidylethanolamine-derived metabolites and isoflavones were identified as potential metabolite biomarkers distinguishing between dietary protein type.
Collapse
Affiliation(s)
- Neil K. Huang
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
| | - Nirupa R. Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
| | - Gregory Matuszek
- Biostatistics and Data Management Unit, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA;
| | - Alice H. Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
| |
Collapse
|
21
|
Perspectives on Mitochondria-ER and Mitochondria-Lipid Droplet Contact in Hepatocytes and Hepatic Lipid Metabolism. Cells 2021; 10:cells10092273. [PMID: 34571924 PMCID: PMC8472694 DOI: 10.3390/cells10092273] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that mitochondrion–endoplasmic reticulum (ER) and mitochondrion–lipid droplet (LD) contact sites are critical in regulating lipid metabolism in cells. It is well established that intracellular organelles communicate with each other continuously through membrane contact sites to maintain organelle function and cellular homeostasis. The accumulation of LDs in hepatocytes is an early indicator of non-alcoholic fatty liver disease (NAFLD) and alcohol-related liver disease (ALD), which may indicate a breakdown in proper inter-organelle communication. In this review, we discuss previous findings in mitochondrion–ER and mitochondrion–LD contact, focusing on their roles in lipid metabolism in hepatocytes. We also present evidence of a unique mitochondrion–LD contact structure in hepatocytes under various physiological and pathological conditions and propose a working hypothesis to speculate about the role of these structures in regulating the functions of mitochondria and LDs and their implications in NAFLD and ALD.
Collapse
|
22
|
Metabolic control of T FH cells and humoral immunity by phosphatidylethanolamine. Nature 2021; 595:724-729. [PMID: 34234346 PMCID: PMC8448202 DOI: 10.1038/s41586-021-03692-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/03/2021] [Indexed: 02/08/2023]
Abstract
T follicular helper (TFH) cells are crucial for B cell-mediated humoral immunity1. Although transcription factors such as BCL6 drive the differentiation of TFH cells2,3, it is unclear whether and how post-transcriptional and metabolic programs enforce TFH cell programming. Here we show that the cytidine diphosphate (CDP)-ethanolamine pathway co-ordinates the expression and localization of CXCR5 with the responses of TFH cells and humoral immunity. Using in vivo CRISPR-Cas9 screening and functional validation in mice, we identify ETNK1, PCYT2, and SELENOI-enzymes in the CDP-ethanolamine pathway for de novo synthesis of phosphatidylethanolamine (PE)-as selective post-transcriptional regulators of TFH cell differentiation that act by promoting the surface expression and functional effects of CXCR5. TFH cells exhibit unique lipid metabolic programs and PE is distributed to the outer layer of the plasma membrane, where it colocalizes with CXCR5. De novo synthesis of PE through the CDP-ethanolamine pathway co-ordinates these events to prevent the internalization and degradation of CXCR5. Genetic deletion of Pcyt2, but not of Pcyt1a (which mediates the CDP-choline pathway), in activated T cells impairs the differentiation of TFH cells, and this is associated with reduced humoral immune responses. Surface levels of PE and CXCR5 expression on B cells also depend on Pcyt2. Our results reveal that phospholipid metabolism orchestrates post-transcriptional mechanisms for TFH cell differentiation and humoral immunity, highlighting the metabolic control of context-dependent immune signalling and effector programs.
Collapse
|
23
|
Chen S, Wu Q, Zhu L, Zong G, Li H, Zheng H, Zeng R, Lin X, Sun L. Plasma glycerophospholipid profile, erythrocyte n-3 PUFAs, and metabolic syndrome incidence: a prospective study in Chinese men and women. Am J Clin Nutr 2021; 114:143-153. [PMID: 33829226 DOI: 10.1093/ajcn/nqab050] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/09/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Animal studies have highlighted critical roles of glycerophospholipid (GP) metabolism in various metabolic syndrome (MetS)-related features such as dyslipidemia, obesity, and insulin resistance. However, human prospective studies of associations between circulating GPs and risks of MetS are scarce. OBJECTIVES We aimed to investigate whether GPs are associated with incidence of MetS in a well-established cohort. METHODS A total of 1243 community-dwelling Chinese aged 50-70 y without MetS at baseline and followed up for 6 y were included in current analyses. A total of 145 plasma GPs were quantified by high-throughput targeted lipidomics. MetS was defined using the updated National Cholesterol Education Program Adult Treatment Panel III criteria for Asian Americans. RESULTS After 6 y, 429 participants developed MetS. Eleven GPs, especially those with long-chain polyunsaturated fatty acids (LCPUFAs) or very-long-chain polyunsaturated fatty acids (VLCPUFAs) at the sn-2 position, including 1 phosphatidylcholine (PC) [PC(18:0/22:6)], 9 phosphatidylethanolamines (PEs) [PE(16:0/22:6), PE(18:0/14:0), PE(18:0/18:1), PE(18:0/18:2), PE(18:0/20:3), PE(18:0/22:5), PE(18:0/22:6), PE(18:1/22:6), and PE(18:2/22:6)], and 1 phosphatidylserine (PS) [PS(18:0/18:0)], were positively associated with incident MetS (RRs: 1.16-1.30 per SD change; Bonferroni-corrected P < 0.05). In network analysis, the strongest positive association for MetS incidence was evidenced in a module mainly composed of PEs containing C22:6 and PSs [RR: 1.21; 95% CI: 1.12, 1.31 per SD change; Bonferroni-corrected P < 0.05]. This association was more pronounced in participants with lower erythrocyte total n-3 PUFA concentrations [Bonferroni-corrected Pinter(P value for the interaction)< 0.05]. CONCLUSIONS Elevated plasma concentrations of GPs, especially PEs with LCPUFAs or VLCPUFAs at the sn-2 position, are associated with higher risk of incident MetS. Future studies are merited to confirm our findings.
Collapse
Affiliation(s)
- Shuangshuang Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingqing Wu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Li Zhu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Geng Zong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huaixing Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - He Zheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Xu Lin
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China.,Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Liang Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
24
|
Taylor A, Grapentine S, Ichhpuniani J, Bakovic M. Choline transporter-like proteins 1 and 2 are newly identified plasma membrane and mitochondrial ethanolamine transporters. J Biol Chem 2021; 296:100604. [PMID: 33789160 PMCID: PMC8081925 DOI: 10.1016/j.jbc.2021.100604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/31/2022] Open
Abstract
The membrane phospholipids phosphatidylcholine and phosphatidylethanolamine (PE) are synthesized de novo by the CDP-choline and CDP-ethanolamine (Kennedy) pathway, in which the extracellular substrates choline and ethanolamine are transported into the cell, phosphorylated, and coupled with diacylglycerol to form the final phospholipid product. Although multiple transport systems have been established for choline, ethanolamine transport is poorly characterized and there is no single protein assigned a transport function for ethanolamine. The solute carriers 44A (SLC44A) known as choline transporter-like proteins-1 and -2 (CTL1 and CTL2) are choline transporter at the plasma membrane and mitochondria. We report a novel function of CTL1 and CTL2 in ethanolamine transport. Using the lack or the gain of gene function in combination with specific antibodies and transport inhibitors we established two distinct ethanolamine transport systems of a high affinity, mediated by CTL1, and of a low affinity, mediated by CTL2. Both transporters are Na+-independent ethanolamine/H+ antiporters. Primary human fibroblasts with separate frameshift mutations in the CTL1 gene (M1= SLC44A1ΔAsp517 and M2= SLC44A1ΔSer126) are devoid of CTL1 ethanolamine transport but maintain unaffected CTL2 transport. The lack of CTL1 in M2 cells reduced the ethanolamine transport, the flux through the CDP-ethanolamine Kennedy pathway, and PE synthesis. In contrast, overexpression of CTL1 in M2 cells improved ethanolamine transport and PE synthesis. These data firmly establish that CTL1 and CTL2 are the first identified ethanolamine transporters in whole cells and mitochondria, with intrinsic roles in de novo PE synthesis by the Kennedy pathway and intracellular redistribution of ethanolamine.
Collapse
Affiliation(s)
- Adrian Taylor
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Sophie Grapentine
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Jasmine Ichhpuniani
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada.
| |
Collapse
|
25
|
Gu X, Al Dubayee M, Alshahrani A, Masood A, Benabdelkamel H, Zahra M, Li L, Abdel Rahman AM, Aljada A. Distinctive Metabolomics Patterns Associated With Insulin Resistance and Type 2 Diabetes Mellitus. Front Mol Biosci 2020; 7:609806. [PMID: 33381523 PMCID: PMC7768025 DOI: 10.3389/fmolb.2020.609806] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/23/2020] [Indexed: 01/17/2023] Open
Abstract
Obesity is associated with an increased risk of insulin resistance (IR) and type 2 diabetes mellitus (T2DM) which is a multi-factorial disease associated with a dysregulated metabolism and can be prevented in pre-diabetic individuals with impaired glucose tolerance. A metabolomic approach emphasizing metabolic pathways is critical to our understanding of this heterogeneous disease. This study aimed to characterize the serum metabolomic fingerprint and multi-metabolite signatures associated with IR and T2DM. Here, we have used untargeted high-performance chemical isotope labeling (CIL) liquid chromatography-mass spectrometry (LC-MS) to identify candidate biomarkers of IR and T2DM in sera from 30 adults of normal weight, 26 obese adults, and 16 adults newly diagnosed with T2DM. Among the 3633 peak pairs detected, 62% were either identified or matched. A group of 78 metabolites were up-regulated and 111 metabolites were down-regulated comparing obese to lean group while 459 metabolites were up-regulated and 166 metabolites were down-regulated comparing T2DM to obese groups. Several metabolites were identified as IR potential biomarkers, including amino acids (Asn, Gln, and His), methionine (Met) sulfoxide, 2-methyl-3-hydroxy-5-formylpyridine-4-carboxylate, serotonin, L-2-amino-3-oxobutanoic acid, and 4,6-dihydroxyquinoline. T2DM was associated with dysregulation of 42 metabolites, including amino acids, amino acids metabolites, and dipeptides. In conclusion, these pilot data have identified IR and T2DM metabolomics panels as potential novel biomarkers of IR and identified metabolites associated with T2DM, with possible diagnostic and therapeutic applications. Further studies to confirm these associations in prospective cohorts are warranted.
Collapse
Affiliation(s)
- Xinyun Gu
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Mohammed Al Dubayee
- Department of Medicine, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Awad Alshahrani
- Department of Medicine, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Afshan Masood
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mahmoud Zahra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Anas M Abdel Rahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Martínez BA, Hoyle RG, Yeudall S, Granade ME, Harris TE, Castle JD, Leitinger N, Bland ML. Innate immune signaling in Drosophila shifts anabolic lipid metabolism from triglyceride storage to phospholipid synthesis to support immune function. PLoS Genet 2020; 16:e1009192. [PMID: 33227003 PMCID: PMC7721134 DOI: 10.1371/journal.pgen.1009192] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 12/07/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
During infection, cellular resources are allocated toward the metabolically-demanding processes of synthesizing and secreting effector proteins that neutralize and kill invading pathogens. In Drosophila, these effectors are antimicrobial peptides (AMPs) that are produced in the fat body, an organ that also serves as a major lipid storage depot. Here we asked how activation of Toll signaling in the larval fat body perturbs lipid homeostasis to understand how cells meet the metabolic demands of the immune response. We find that genetic or physiological activation of fat body Toll signaling leads to a tissue-autonomous reduction in triglyceride storage that is paralleled by decreased transcript levels of the DGAT homolog midway, which carries out the final step of triglyceride synthesis. In contrast, Kennedy pathway enzymes that synthesize membrane phospholipids are induced. Mass spectrometry analysis revealed elevated levels of major phosphatidylcholine and phosphatidylethanolamine species in fat bodies with active Toll signaling. The ER stress mediator Xbp1 contributed to the Toll-dependent induction of Kennedy pathway enzymes, which was blunted by deleting AMP genes, thereby reducing secretory demand elicited by Toll activation. Consistent with ER stress induction, ER volume is expanded in fat body cells with active Toll signaling, as determined by transmission electron microscopy. A major functional consequence of reduced Kennedy pathway induction is an impaired immune response to bacterial infection. Our results establish that Toll signaling induces a shift in anabolic lipid metabolism to favor phospholipid synthesis and ER expansion that may serve the immediate demand for AMP synthesis and secretion but with the long-term consequence of insufficient nutrient storage.
Collapse
Affiliation(s)
- Brittany A. Martínez
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA, United States of America
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
| | - Rosalie G. Hoyle
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
| | - Scott Yeudall
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA, United States of America
| | - Mitchell E. Granade
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA, United States of America
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
| | - Thurl E. Harris
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA, United States of America
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
| | - J. David Castle
- Department of Cell Biology, University of Virginia, Charlottesville, VA, United States of America
| | - Norbert Leitinger
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA, United States of America
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
| | - Michelle L. Bland
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA, United States of America
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States of America
- * E-mail:
| |
Collapse
|
27
|
Zhao H, Wang T. PE homeostasis rebalanced through mitochondria-ER lipid exchange prevents retinal degeneration in Drosophila. PLoS Genet 2020; 16:e1009070. [PMID: 33064773 PMCID: PMC7592913 DOI: 10.1371/journal.pgen.1009070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/28/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
The major glycerophospholipid phosphatidylethanolamine (PE) in the nervous system is essential for neural development and function. There are two major PE synthesis pathways, the CDP-ethanolamine pathway in the endoplasmic reticulum (ER) and the phosphatidylserine decarboxylase (PSD) pathway in mitochondria. However, the role played by mitochondrial PE synthesis in maintaining cellular PE homeostasis is unknown. Here, we show that Drosophila pect (phosphoethanolamine cytidylyltransferase) mutants lacking the CDP-ethanolamine pathway, exhibited alterations in phospholipid composition, defective phototransduction, and retinal degeneration. Induction of the PSD pathway fully restored levels and composition of cellular PE, thus rescued the retinal degeneration and defective visual responses in pect mutants. Disrupting lipid exchange between mitochondria and ER blocked the ability of PSD to rescue pect mutant phenotypes. These findings provide direct evidence that the synthesis of PE in mitochondria contributes to cellular PE homeostasis, and suggest the induction of mitochondrial PE synthesis as a promising therapeutic approach for disorders associated with PE deficiency.
Collapse
Affiliation(s)
- Haifang Zhao
- National Institute of Biological Sciences, Beijing, China
| | - Tao Wang
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
28
|
Rescue of Hepatic Phospholipid Remodeling Defectin iPLA2β-Null Mice Attenuates Obese but Not Non-Obese Fatty Liver. Biomolecules 2020; 10:biom10091332. [PMID: 32957701 PMCID: PMC7565968 DOI: 10.3390/biom10091332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022] Open
Abstract
Polymorphisms of group VIA calcium-independent phospholipase A2 (iPLA2β or PLA2G6) are positively associated with adiposity, blood lipids, and Type-2 diabetes. The ubiquitously expressed iPLA2β catalyzes the hydrolysis of phospholipids (PLs) to generate a fatty acid and a lysoPL. We studied the role of iPLA2β on PL metabolism in non-alcoholic fatty liver disease (NAFLD). By using global deletion iPLA2β-null mice, we investigated three NAFLD mouse models; genetic Ob/Ob and long-term high-fat-diet (HFD) feeding (representing obese NAFLD) as well as feeding with methionine- and choline-deficient (MCD) diet (representing non-obese NAFLD). A decrease of hepatic PLs containing monounsaturated- and polyunsaturated fatty acids and a decrease of the ratio between PLs and cholesterol esters were observed in all three NAFLD models. iPLA2β deficiency rescued these decreases in obese, but not in non-obese, NAFLD models. iPLA2β deficiency elicited protection against fatty liver and obesity in the order of Ob/Ob › HFD » MCD. Liver inflammation was not protected in HFD NAFLD, and that liver fibrosis was even exaggerated in non-obese MCD model. Thus, the rescue of hepatic PL remodeling defect observed in iPLA2β-null mice was critical for the protection against NAFLD and obesity. However, iPLA2β deletion in specific cell types such as macrophages may render liver inflammation and fibrosis, independent of steatosis protection.
Collapse
|
29
|
Bauer KC, Huus KE, Brown EM, Bozorgmehr T, Petersen C, Cirstea MS, Woodward SE, McCoy J, Hun J, Pamplona R, Ayala V, Finlay BB. Dietary Intervention Reverses Fatty Liver and Altered Gut Microbiota during Early-Life Undernutrition. mSystems 2020; 5:e00499-20. [PMID: 32900869 PMCID: PMC7483509 DOI: 10.1128/msystems.00499-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/01/2020] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), largely studied as a condition of overnutrition, also presents in undernourished populations. Like NAFLD, undernutrition disrupts systemic metabolism and has been linked to gut microbiota dysbiosis. Indeed, chronic exposures to fecal microbes contribute to undernutrition pathology in regions with poor sanitation. Despite a growing prevalence of fatty liver disease, the influence of undernutrition and the gut microbiota remain largely unexplored. Here, we utilize an established murine model (C57BL/6J mice placed on a malnourished diet that received iterative Escherichia coli/Bacteroidales gavage [MBG mice]) that combines a protein/fat-deficient diet and iterative exposure to specific, fecal microbes. Fecal-oral contamination exacerbates triglyceride accumulation in undernourished mice. MBG livers exhibit diffuse lipidosis accompanied by striking shifts in fatty acid, glycerophospholipid, and retinol metabolism. Multiomic analyses revealed metabolomic pathways linked to the undernourished gut microbiome and hepatic steatosis, including phenylacetate metabolism. Intriguingly, fatty liver features were observed only in the early-life, but not adult, MBG model despite similar liver metabolomic profiles. Importantly, we demonstrate that dietary intervention largely mitigates aberrant metabolomic and microbiome features in MBG mice. These findings indicate a crucial window in early-life development that, when disrupted by nutritional deficiency, may significantly influence liver function. Our work provides a multifaceted study of how diet and gut microbes inform fatty liver progression and reversal during undernutrition.IMPORTANCE Nonalcoholic fatty liver disease (NAFLD) remains a global epidemic, but it is often studied in the context of obesity and aging. Nutritional deficits, however, also trigger hepatic steatosis, influencing health trajectories in undernourished pediatric populations. Here, we report that exposure to specific gut microbes impacts fatty liver pathology in mice fed a protein/fat-deficient diet. We utilize a multiomics approach to (i) characterize NAFLD in the context of early undernutrition and (ii) examine the impact of diet and gut microbes in the pathology and reversal of hepatic steatosis. We provide compelling evidence that an early-life, critical development window facilitates undernutrition-induced fatty liver pathology. Moreover, we demonstrate that sustained dietary intervention largely reverses fatty liver features and microbiome shifts observed during early-life malnutrition.
Collapse
Affiliation(s)
- K C Bauer
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - K E Huus
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - E M Brown
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - T Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Petersen
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - M S Cirstea
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - S E Woodward
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - J McCoy
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Hun
- The Metabolomics Innovation Centre, University of Victoria, British Columbia, Canada
| | - R Pamplona
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain
- Department of Metabolomic Physiology, Universitat de Lleida, Lleida, Spain
| | - V Ayala
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain
- Department of Metabolomic Physiology, Universitat de Lleida, Lleida, Spain
| | - B B Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Microbiology and Immunology Department, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, Canada
| |
Collapse
|
30
|
Gliniak CM, Scherer PE. PHOSPHO1 puts the breaks on thermogenesis in brown adipocytes. Proc Natl Acad Sci U S A 2020; 117:16726-16728. [PMID: 32641502 PMCID: PMC7382279 DOI: 10.1073/pnas.2011052117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Christy M Gliniak
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549;
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549
| |
Collapse
|
31
|
Santinha D, Klopot A, Marques I, Ellis E, Jorns C, Johansson H, Melo T, Antonson P, Jakobsson T, Félix V, Gustafsson JÅ, Domingues MR, Mode A, Helguero LA. Lipidomic analysis of human primary hepatocytes following LXR activation with GW3965 identifies AGXT2L1 as a main target associated to changes in phosphatidylethanolamine. J Steroid Biochem Mol Biol 2020; 198:105558. [PMID: 31783151 DOI: 10.1016/j.jsbmb.2019.105558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023]
Abstract
Liver X receptor (LXR) agonists have the potential to alleviate obesity related diseases, particularly atherosclerosis. However, LXRs are transcriptional regulators that induce de novo lipogenesis and lipid accumulation in hepatocytes which represents a serious adverse effect. In this work, we sought to characterize the LXR agonist GW3965 effects on fatty acid (FA) and phospholipid (PL) remodelling and the correlation with gene expression in order to better understand the underlying effects leading to hepatic pathology upon LXR activation. Human primary hepatocytes treated for 48 h with GW3965 were analysed for changes in lipid metabolism gene expression by qPCR, variations in the FA profile was evaluated by GC-FID and in PL profiles using thin layer chromatography, ESI-MS and MS/MS analysis. Changes in cell membrane biochemical properties were studied using bilayer models generated with CHARMM-GUI. ELOLV6 and SCD1 mRNA increase was consistent with higher C16:1 and C18:1n9 at the expense of C16:0 and C18:0. The reduction of C18:2n6 and increase in C20:2n6 was in agreement with ELOVL5 upregulation. Phosphatydilethanolamine (PE) levels tended to decrease and phosphatidylinositol to increase; although differences did not reach significance, they correlated with changes in AGXT2L1, CDS1 and LPIN1 mRNA levels that were increased. The overall effect of GW3965 on PEs molecular profiles was an increase of long-chain polyunsaturated FA chains and a decrease of C16/C18 saturated and monounsaturated FAs chains. Additionally, PC (32:1) and PC (34:2) were decreased, and PC (36:1) and PC (34:1) were increased. AGXT2L1 is an enzyme with strict substrate specificity for phosphoethanolamine, which is converted into ammonia in GW3965-treated hepatocytes and could explain the PE reduction. In summary, LXR activation by GW3965 targets PE biosynthesis and FA elongation/desaturation, which tends to decrease PE in relation to total PL levels, and remodelling of PC and PE molecular species. We identified the human AGXT2L1 gene as induced by LXR activation by both synthetic and endogenous agonist treatment. The increase in acetaldehyde-induced oxidative stress, and in the lipid species identified have the potential to enhance the inflammatory process and impair membrane function. Future studies should focus on inhibition of AGXT2L1 activity with the aim of reverting the steatosis induced by LXR activation.
Collapse
Affiliation(s)
- Deolinda Santinha
- Department of Chemistry, QOPNA Research Unit, University of Aveiro, Portugal
| | - Anna Klopot
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Huddinge, Sweden
| | - Igor Marques
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Unit for Liver Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Carl Jorns
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Unit for Liver Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Helene Johansson
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Transplantation Surgery, Unit for Liver Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tânia Melo
- Department of Chemistry, QOPNA Research Unit, University of Aveiro, Portugal; Department of Chemistry, CESAM&ECOMARE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Per Antonson
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Huddinge, Sweden
| | - Tomas Jakobsson
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Vítor Félix
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Huddinge, Sweden; Center for Nuclear Receptors and Cell Signaling, Department of Cell Biology and Biochemistry, University of Houston, TX, United States
| | - Maria Rosário Domingues
- Department of Chemistry, QOPNA Research Unit, University of Aveiro, Portugal; Department of Chemistry, CESAM&ECOMARE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Agneta Mode
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Huddinge, Sweden
| | - Luisa A Helguero
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
32
|
Guan Y, Chen X, Wu M, Zhu W, Arslan A, Takeda S, Nguyen MH, Majeti R, Thomas D, Zheng M, Peltz G. The phosphatidylethanolamine biosynthesis pathway provides a new target for cancer chemotherapy. J Hepatol 2020; 72:746-760. [PMID: 31760071 PMCID: PMC7085447 DOI: 10.1016/j.jhep.2019.11.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Since human induced pluripotent stem cells (iPSCs) develop into hepatic organoids through stages that resemble human embryonic liver development, they can be used to study developmental processes and disease pathology. Therefore, we examined the early stages of hepatic organoid formation to identify key pathways affecting early liver development. METHODS Single-cell RNA-sequencing and metabolomic analysis was performed on developing organoid cultures at the iPSC, hepatoblast (day 9) and mature organoid stage. The importance of the phosphatidylethanolamine biosynthesis pathway to early liver development was examined in developing organoid cultures using iPSC with a CRISPR-mediated gene knockout and an over the counter medication (meclizine) that inhibits the rate-limiting enzyme in this pathway. Meclizine's effect on the growth of a human hepatocarcinoma cell line in a xenotransplantation model and on the growth of acute myeloid leukemia cells in vitro was also examined. RESULTS Transcriptomic and metabolomic analysis of organoid development indicated that the phosphatidylethanolamine biosynthesis pathway is essential for early liver development. Unexpectedly, early hepatoblasts were selectively sensitive to the cytotoxic effect of meclizine. We demonstrate that meclizine could be repurposed for use in a new synergistic combination therapy for primary liver cancer: a glycolysis inhibitor reprograms cancer cell metabolism to make it susceptible to the cytotoxic effect of meclizine. This combination inhibited the growth of a human liver carcinoma cell line in vitro and in a xenotransplantation model, without causing significant side effects. This drug combination was also highly active against acute myeloid leukemia cells. CONCLUSION Our data indicate that phosphatidylethanolamine biosynthesis is a targetable pathway for cancer; meclizine may have clinical efficacy as a repurposed anti-cancer drug when used as part of a new combination therapy. LAY SUMMARY The early stages of human liver development were modeled using human hepatic organoids. We identified a pathway that was essential for early liver development. Based upon this finding, a novel combination drug therapy was identified that could be used to treat primary liver cancer and possibly other types of cancer.
Collapse
Affiliation(s)
- Yuan Guan
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Xinyu Chen
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Manhong Wu
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Wan Zhu
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Ahmed Arslan
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Saori Takeda
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Mindie H. Nguyen
- Department of Medicine, Division of Gastroenterology and
Hepatology, Stanford University School of Medicine, Stanford CA 94305
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer
Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford
University School of Medicine
| | - Dan Thomas
- Department of Medicine, Division of Hematology, Cancer
Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford
University School of Medicine
| | - Ming Zheng
- Department of Anesthesia, Stanford University School of
Medicine, Stanford CA 94305
| | - Gary Peltz
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305.
| |
Collapse
|
33
|
Phosphatidylserine decarboxylase is critical for the maintenance of skeletal muscle mitochondrial integrity and muscle mass. Mol Metab 2019; 27:33-46. [PMID: 31285171 PMCID: PMC6717954 DOI: 10.1016/j.molmet.2019.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/12/2019] [Accepted: 06/23/2019] [Indexed: 11/24/2022] Open
Abstract
Objective Phosphatidylethanolamine (PtdEtn) is a major phospholipid in mammals. It is synthesized via two pathways, the CDP-ethanolamine pathway in the endoplasmic reticulum and the phosphatidylserine (PtdSer) decarboxylase (PSD) pathway in the mitochondria. While the CDP-ethanolamine pathway is considered the major route for PtdEtn synthesis in most mammalian tissues, little is known about the importance of the PSD pathway in vivo, especially in tissues enriched with mitochondria such as skeletal muscle. Therefore, we aimed to examine the role of the mitochondrial PSD pathway in regulating PtdEtn homeostasis in skeletal muscle in vivo. Methods To determine the functional significance of this pathway in skeletal muscle in vivo, an adeno-associated viral vector approach was employed to knockdown PSD expression in skeletal muscle of adult mice. Muscle lipid and metabolite profiling was performed using mass spectrometry. Results PSD knockdown disrupted muscle phospholipid homeostasis leading to an ∼25% reduction in PtdEtn and an ∼45% increase in PtdSer content. This was accompanied by the development of a severe myopathy, evident by a 40% loss in muscle mass as well as extensive myofiber damage as shown by increased DNA synthesis and central nucleation. In addition, PSD knockdown caused marked accumulation of abnormally appearing mitochondria that exhibited severely disrupted inner membrane integrity and reduced OXPHOS protein content. Conclusions The PSD pathway has a significant role in maintaining phospholipid homeostasis in adult skeletal muscle. Moreover, PSD is essential for maintenance of mitochondrial integrity and skeletal muscle mass. The PSD pathway has an important role in regulating muscle phospholipid homeostasis. Disrupting the PSD pathway caused marked muscle wasting and myofibre damage. Knockdown of PSD caused accumulation of mitochondria with ultrastructural defects. PSD is important in regulating mitochondrial integrity and skeletal muscle mass.
Collapse
|
34
|
Lee J, Ridgway ND. Substrate channeling in the glycerol-3-phosphate pathway regulates the synthesis, storage and secretion of glycerolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158438. [PMID: 30959116 DOI: 10.1016/j.bbalip.2019.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/16/2023]
Abstract
The successive acylation of glycerol-3-phosphate (G3P) by glycerol-3-phosphate acyltransferases and acylglycerol-3-phosphate acyltransferases produces phosphatidic acid (PA), a precursor for CDP-diacylglycerol-dependent phospholipid synthesis. PA is further dephosphorylated by LIPINs to produce diacylglycerol (DG), a substrate for the synthesis of triglyceride (TG) by DG acyltransferases and a precursor for phospholipid synthesis via the CDP-choline and CDP-ethanolamine (Kennedy) pathways. The channeling of fatty acids into TG for storage in lipid droplets and secretion in lipoproteins or phospholipids for membrane biogenesis is dependent on isoform expression, activity and localization of G3P pathway enzymes, as well as dietary and hormonal and tissue-specific factors. Here, we review the mechanisms that control partitioning of substrates into lipid products of the G3P pathway.
Collapse
Affiliation(s)
- Jonghwa Lee
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Neale D Ridgway
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
35
|
Argov-Argaman N. Symposium review: Milk fat globule size: Practical implications and metabolic regulation. J Dairy Sci 2019; 102:2783-2795. [PMID: 30639008 DOI: 10.3168/jds.2018-15240] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Milk fat globule (MFG) size ranges over 3 orders of magnitude, from less than 200 nm to over 15 µm. The significance of MFG size derives from its tight association with its lipidome and proteome. More specifically, small MFG have relatively higher content of membrane compared with large globules, and this membrane exerts diverse positive health effects, as reported in human and animal studies. In addition, MFG size has industrial significance, as it affects the physicochemical and sensory characteristics of dairy products. Studies on the size regulation of MFG are scarce, mainly because various confounders indirectly affect MFG size. Because MFG size is determined before and during its secretion from mammary epithelial cells, studies on the size regulation of its precursors, the intracellular lipid droplets (LD), have been used as a proxy for understanding the mechanisms controlling MFG size. In this review, we provide evidence for 2 distinct mechanisms regulating LD size in mammary epithelial cells: co-regulation of fat content and triglyceride-synthesis capacity of the cells, and fusion between LD. The latter is controlled by the membrane's polar lipid composition and involves mitochondrial enzymes. Accordingly, this review also discusses MFG size regulation in the in vivo metabolic context, as MFG morphometric features are often modulated under conditions that involve animals' altered energy status.
Collapse
Affiliation(s)
- Nurit Argov-Argaman
- Department of Animal Science, the Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Israel, POB 76100.
| |
Collapse
|
36
|
Weber K, Casali C, Gaveglio V, Pasquaré S, Morel Gómez E, Parra L, Erjavec L, Perazzo C, Fernández Tome MC. TAG synthesis and storage under osmotic stress. A requirement for preserving membrane homeostasis in renal cells. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1108-1120. [DOI: 10.1016/j.bbalip.2018.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/11/2018] [Accepted: 06/12/2018] [Indexed: 12/14/2022]
|
37
|
Snider SA, Margison KD, Ghorbani P, LeBlond ND, O'Dwyer C, Nunes JRC, Nguyen T, Xu H, Bennett SAL, Fullerton MD. Choline transport links macrophage phospholipid metabolism and inflammation. J Biol Chem 2018; 293:11600-11611. [PMID: 29880645 DOI: 10.1074/jbc.ra118.003180] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
Choline is an essential nutrient that is required for synthesis of the main eukaryote phospholipid, phosphatidylcholine. Macrophages are innate immune cells that survey and respond to danger and damage signals. Although it is well-known that energy metabolism can dictate macrophage function, little is known as to the importance of choline homeostasis in macrophage biology. We hypothesized that the uptake and metabolism of choline are important for macrophage inflammation. Polarization of primary bone marrow macrophages with lipopolysaccharide (LPS) resulted in an increased rate of choline uptake and higher levels of PC synthesis. This was attributed to a substantial increase in the transcript and protein expression of the choline transporter-like protein-1 (CTL1) in polarized cells. We next sought to determine the importance of choline uptake and CTL1 for macrophage immune responsiveness. Chronic pharmacological or CTL1 antibody-mediated inhibition of choline uptake resulted in altered cytokine secretion in response to LPS, which was associated with increased levels of diacylglycerol and activation of protein kinase C. These experiments establish a previously unappreciated link between choline phospholipid metabolism and macrophage immune responsiveness, highlighting a critical and regulatory role for macrophage choline uptake via the CTL1 transporter.
Collapse
Affiliation(s)
- Shayne A Snider
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kaitlyn D Margison
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Peyman Ghorbani
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Nicholas D LeBlond
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Conor O'Dwyer
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Julia R C Nunes
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Thao Nguyen
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; the Ottawa Institute of Systems Biology and University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Hongbin Xu
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; the Ottawa Institute of Systems Biology and University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Steffany A L Bennett
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; the Ottawa Institute of Systems Biology and University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Morgan D Fullerton
- University of Ottawa Centre for Infection, Immunity, and Inflammation and Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
38
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a group of liver disorders encompassing simple hepatic steatosis and its more aggressive forms of nonalcoholic steatohepatitis and cirrhosis. It is a rapidly growing health concern and the major cause for the increasing incidence of primary liver tumors. Unequivocal evidence shows that sphingolipid metabolism is altered in the course of the disease and these changes might contribute to NAFLD progression. Recent data provide solid support to the notion that deregulated ceramide and sphingosine-1-phosphate metabolism are present at all stages of NAFLD, i.e., steatosis, nonalcoholic steatohepatitis, advanced fibrosis, and hepatocellular carcinoma (HCC). Insulin sensitivity, de novo lipogenesis, and the resulting lipotoxicity, fibrosis, and angiogenesis are all seemingly regulated in a manner that involves either ceramide and/or sphingosine-1-phosphate. Sphingolipids might also participate in the onset of hepatocellular senescence. The latter has been shown to contribute to the advancement of cirrhosis to HCC in the classical cases of end-stage liver disease, i.e., viral- or alcohol-induced; however, emerging evidence suggests that senescence is also involved in the pathogenicity of NAFLD possibly via changes in ceramide metabolism.
Collapse
|
39
|
Vance JE. Historical perspective: phosphatidylserine and phosphatidylethanolamine from the 1800s to the present. J Lipid Res 2018; 59:923-944. [PMID: 29661786 DOI: 10.1194/jlr.r084004] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/12/2018] [Indexed: 12/17/2022] Open
Abstract
This article provides a historical account of the discovery, chemistry, and biochemistry of two ubiquitous phosphoglycerolipids, phosphatidylserine (PS) and phosphatidylethanolamine (PE), including the ether lipids. In addition, the article describes the biosynthetic pathways for these phospholipids and how these pathways were elucidated. Several unique functions of PS and PE in mammalian cells in addition to their ability to define physical properties of membranes are discussed. For example, the translocation of PS from the inner to the outer leaflet of the plasma membrane of cells occurs during apoptosis and during some other specific physiological processes, and this translocation is responsible for profound life-or-death events. Moreover, mitochondrial function is severely impaired when the PE content of mitochondria is reduced below a threshold level. The discovery and implications of the existence of membrane contact sites between the endoplasmic reticulum and mitochondria and their relevance for PS and PE metabolism, as well as for mitochondrial function, are also discussed. Many of the recent advances in these fields are due to the use of isotope labeling for tracing biochemical pathways. In addition, techniques for disruption of specific genes in mice are now widely used and have provided major breakthroughs in understanding the roles and metabolism of PS and PE in vivo.
Collapse
Affiliation(s)
- Jean E Vance
- Department of Medicine and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2S2, Canada.
| |
Collapse
|
40
|
Ho N, Xu C, Thibault G. From the unfolded protein response to metabolic diseases - lipids under the spotlight. J Cell Sci 2018; 131:131/3/jcs199307. [PMID: 29439157 DOI: 10.1242/jcs.199307] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The unfolded protein response (UPR) is classically viewed as a stress response pathway to maintain protein homeostasis at the endoplasmic reticulum (ER). However, it has recently emerged that the UPR can be directly activated by lipid perturbation, independently of misfolded proteins. Comprising primarily phospholipids, sphingolipids and sterols, individual membranes can contain hundreds of distinct lipids. Even with such complexity, lipid distribution in a cell is tightly regulated by mechanisms that remain incompletely understood. It is therefore unsurprising that lipid dysregulation can be a key factor in disease development. Recent advances in analysis of lipids and their regulators have revealed remarkable mechanisms and connections to other cellular pathways including the UPR. In this Review, we summarize the current understanding in UPR transducers functioning as lipid sensors and the interplay between lipid metabolism and ER homeostasis in the context of metabolic diseases. We attempt to provide a framework consisting of a few key principles to integrate the different lines of evidence and explain this rather complicated mechanism.
Collapse
Affiliation(s)
- Nurulain Ho
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| | - Chengchao Xu
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142-1479, USA
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551
| |
Collapse
|
41
|
Cohen BC, Raz C, Shamay A, Argov-Argaman N. Lipid Droplet Fusion in Mammary Epithelial Cells is Regulated by Phosphatidylethanolamine Metabolism. J Mammary Gland Biol Neoplasia 2017; 22:235-249. [PMID: 29188493 DOI: 10.1007/s10911-017-9386-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Mammary epithelial cells (MEC) secrete fat in the form of milk fat globules (MFG) which are found in milk in diverse sizes. MFG originate from intracellular lipid droplets, and the mechanism underlying their size regulation is still elusive. Two main mechanisms have been suggested to control lipid droplet size. The first is a well-documented pathway, which involves regulation of cellular triglyceride content. The second is the fusion pathway, which is less-documented, especially in mammalian cells, and its importance in the regulation of droplet size is still unclear. Using biochemical and molecular inhibitors, we provide evidence that in MEC, lipid droplet size is determined by fusion, independent of cellular triglyceride content. The extent of fusion is determined by the cell membrane's phospholipid composition. In particular, increasing phosphatidylethanolamine (PE) content enhances fusion between lipid droplets and hence increases lipid droplet size. We further identified the underlying biochemical mechanism that controls this content as the mitochondrial enzyme phosphatidylserine decarboxylase; siRNA knockdown of this enzyme reduced the number of large lipid droplets threefold. Further, inhibition of phosphatidylserine transfer to the mitochondria, where its conversion to PE occurs, diminished the large lipid droplet phenotype in these cells. These results reveal, for the first time to our knowledge in mammalian cells and specifically in mammary epithelium, the missing biochemical link between the metabolism of cellular complex lipids and lipid-droplet fusion, which ultimately defines lipid droplet size.
Collapse
Affiliation(s)
- Bat-Chen Cohen
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel.
| | - Chen Raz
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| | - Avi Shamay
- Department of Ruminant Science, Agricultural Research Organization, Volcani Center, Bet Dagan, Israel
| | - Nurit Argov-Argaman
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| |
Collapse
|
42
|
Zhang Y, Udayakumar D, Cai L, Hu Z, Kapur P, Kho EY, Pavía-Jiménez A, Fulkerson M, de Leon AD, Yuan Q, Dimitrov IE, Yokoo T, Ye J, Mitsche MA, Kim H, McDonald JG, Xi Y, Madhuranthakam AJ, Dwivedi DK, Lenkinski RE, Cadeddu JA, Margulis V, Brugarolas J, DeBerardinis RJ, Pedrosa I. Addressing metabolic heterogeneity in clear cell renal cell carcinoma with quantitative Dixon MRI. JCI Insight 2017; 2:94278. [PMID: 28768909 DOI: 10.1172/jci.insight.94278] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dysregulated lipid and glucose metabolism in clear cell renal cell carcinoma (ccRCC) has been implicated in disease progression, and whole tumor tissue-based assessment of these changes is challenged by the tumor heterogeneity. We studied a noninvasive quantitative MRI method that predicts metabolic alterations in the whole tumor. METHODS We applied Dixon-based MRI for in vivo quantification of lipid accumulation (fat fraction [FF]) in targeted regions of interest of 45 primary ccRCCs and correlated these MRI measures to mass spectrometry-based lipidomics and metabolomics of anatomically colocalized tissue samples isolated from the same tumor after surgery. RESULTS In vivo tumor FF showed statistically significant (P < 0.0001) positive correlation with histologic fat content (Spearman correlation coefficient, ρ = 0.79), spectrometric triglycerides (ρ = 0.56) and cholesterol (ρ = 0.47); it showed negative correlation with free fatty acids (ρ = -0.44) and phospholipids (ρ = -0.65). We observed both inter- and intratumoral heterogeneity in lipid accumulation within the same tumor grade, whereas most aggressive tumors (International Society of Urological Pathology [ISUP] grade 4) exhibited reduced lipid accumulation. Cellular metabolites in tumors were altered compared with adjacent renal parenchyma. CONCLUSION Our results support the use of noninvasive quantitative Dixon-based MRI as a biomarker of reprogrammed lipid metabolism in ccRCC, which may serve as a predictor of tumor aggressiveness before surgical intervention. FUNDING NIH R01CA154475 (YZ, MF, PK, IP), NIH P50CA196516 (IP, JB, RJD, JAC, PK), Welch Foundation I-1832 (JY), and NIH P01HL020948 (JGM).
Collapse
Affiliation(s)
| | | | - Ling Cai
- Children's Medical Center Research Institute.,Quantitative Biomedical Research Center
| | - Zeping Hu
- Children's Medical Center Research Institute
| | - Payal Kapur
- Pathology.,Urology.,Kidney Cancer Program - Simmons Comprehensive Cancer Center, and
| | | | - Andrea Pavía-Jiménez
- Kidney Cancer Program - Simmons Comprehensive Cancer Center, and.,Internal Medicine, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | - Ivan E Dimitrov
- Advanced Imaging Research Center.,Philips Medical Systems, Cleveland, Ohio, USA
| | | | - Jin Ye
- Molecular Genetics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew A Mitsche
- Molecular Genetics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hyeonwoo Kim
- Molecular Genetics, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | - James Brugarolas
- Kidney Cancer Program - Simmons Comprehensive Cancer Center, and.,Internal Medicine, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | | | - Ivan Pedrosa
- Radiology.,Advanced Imaging Research Center.,Kidney Cancer Program - Simmons Comprehensive Cancer Center, and
| |
Collapse
|
43
|
The expanding role of sphingolipids in lipid droplet biogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1155-1165. [PMID: 28743537 DOI: 10.1016/j.bbalip.2017.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/17/2023]
Abstract
Sphingolipids are a diverse class of lipids that have regulatory, structural, and metabolic functions. Although chemically distinct from the neutral lipids and the glycerophospholipids, which are the main lipid components of the lipid droplets, sphingolipids have nonetheless been shown to influence lipid droplet formation. The goal of this article is to review the available information and provide a cohesive picture of the role sphingolipids play in lipid droplet biogenesis. The following topics are discussed: (i) the abundance of sphingolipids in lipid droplets and their functional significance; (ii) cross-talk between the synthetic pathways of sphingolipids, glycerophospholipids, and neutral lipids; (iii) the impact of bioactive sphingolipids on TAG synthesis and degradation; (iv) interactions between sphingolipids and other lipid droplet components, like cholesterol esters and proteins; (v) inhibition/genetic deletion of specific sphingolipid metabolic enzymes and the resulting effects on lipid droplet formation in mouse models. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
|
44
|
Ethanolamine and Phosphatidylethanolamine: Partners in Health and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4829180. [PMID: 28785375 PMCID: PMC5529665 DOI: 10.1155/2017/4829180] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022]
Abstract
Phosphatidylethanolamine (PE) is the second most abundant phospholipid in mammalian cells. PE comprises about 15–25% of the total lipid in mammalian cells; it is enriched in the inner leaflet of membranes, and it is especially abundant in the inner mitochondrial membrane. PE has quite remarkable activities: it is a lipid chaperone that assists in the folding of certain membrane proteins, it is required for the activity of several of the respiratory complexes, and it plays a key role in the initiation of autophagy. In this review, we focus on PE's roles in lipid-induced stress in the endoplasmic reticulum (ER), Parkinson's disease (PD), ferroptosis, and cancer.
Collapse
|
45
|
van der Veen JN, Kennelly JP, Wan S, Vance JE, Vance DE, Jacobs RL. The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1558-1572. [PMID: 28411170 DOI: 10.1016/j.bbamem.2017.04.006] [Citation(s) in RCA: 937] [Impact Index Per Article: 117.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/27/2017] [Accepted: 04/09/2017] [Indexed: 12/11/2022]
Abstract
Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) are the most abundant phospholipids in all mammalian cell membranes. In the 1950s, Eugene Kennedy and co-workers performed groundbreaking research that established the general outline of many of the pathways of phospholipid biosynthesis. In recent years, the importance of phospholipid metabolism in regulating lipid, lipoprotein and whole-body energy metabolism has been demonstrated in numerous dietary studies and knockout animal models. The purpose of this review is to highlight the unappreciated impact of phospholipid metabolism on health and disease. Abnormally high, and abnormally low, cellular PC/PE molar ratios in various tissues can influence energy metabolism and have been linked to disease progression. For example, inhibition of hepatic PC synthesis impairs very low density lipoprotein secretion and changes in hepatic phospholipid composition have been linked to fatty liver disease and impaired liver regeneration after surgery. The relative abundance of PC and PE regulates the size and dynamics of lipid droplets. In mitochondria, changes in the PC/PE molar ratio affect energy production. We highlight data showing that changes in the PC and/or PE content of various tissues are implicated in metabolic disorders such as atherosclerosis, insulin resistance and obesity. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Jelske N van der Veen
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - John P Kennelly
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Agricultural, Food and Nutritional Science, 4-002 Li Ka Shing Centre for Heath Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Sereana Wan
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Jean E Vance
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Medicine, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - René L Jacobs
- Group on the Molecular and Cell Biology of Lipids, Canada; Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; Department of Agricultural, Food and Nutritional Science, 4-002 Li Ka Shing Centre for Heath Research Innovations, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
46
|
Deevska GM, Dotson PP, Karakashian AA, Isaac G, Wrona M, Kelly SB, Merrill AH, Nikolova-Karakashian MN. Novel Interconnections in Lipid Metabolism Revealed by Overexpression of Sphingomyelin Synthase-1. J Biol Chem 2017; 292:5110-5122. [PMID: 28087695 DOI: 10.1074/jbc.m116.751602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 01/13/2017] [Indexed: 12/11/2022] Open
Abstract
This study investigates the consequences of elevating sphingomyelin synthase 1 (SMS1) activity, which generates the main mammalian sphingolipid, sphingomyelin. HepG2 cells stably transfected with SMS1 (HepG2-SMS1) exhibit elevated enzyme activity in vitro and increased sphingomyelin content (mainly C22:0- and C24:0-sphingomyelin) but lower hexosylceramide (Hex-Cer) levels. HepG2-SMS1 cells have fewer triacylglycerols than controls but similar diacylglycerol acyltransferase activity, triacylglycerol secretion, and mitochondrial function. Treatment with 1 mm palmitate increases de novo ceramide synthesis in both cell lines to a similar degree, causing accumulation of C16:0-ceramide (and some C18:0-, C20:0-, and C22:0-ceramides) as well as C16:0- and C18:0-Hex-Cers. In these experiments, the palmitic acid is delivered as a complex with delipidated BSA (2:1, mol/mol) and does not induce significant lipotoxicity. Based on precursor labeling, the flux through SM synthase also increases, which is exacerbated in HepG2-SMS1 cells. In contrast, palmitate-induced lipid droplet formation is significantly reduced in HepG2-SMS1 cells. [14C]Choline and [3H]palmitate tracking shows that SMS1 overexpression apparently affects the partitioning of palmitate-enriched diacylglycerol between the phosphatidylcholine and triacylglycerol pathways, to the benefit of the former. Furthermore, triacylglycerols from HepG2-SMS1 cells are enriched in polyunsaturated fatty acids, which is indicative of active remodeling. Together, these results delineate novel metabolic interactions between glycerolipids and sphingolipids.
Collapse
Affiliation(s)
- Gergana M Deevska
- From the Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | - Patrick P Dotson
- From the Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | | | - Giorgis Isaac
- Pharmaceutical Discovery and Life Sciences, Waters Corporation, Milford, Massachusetts 01757, and
| | - Mark Wrona
- Pharmaceutical Discovery and Life Sciences, Waters Corporation, Milford, Massachusetts 01757, and
| | - Samuel B Kelly
- the School of Biology and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Alfred H Merrill
- the School of Biology and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332
| | | |
Collapse
|
47
|
Calzada E, Onguka O, Claypool SM. Phosphatidylethanolamine Metabolism in Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 321:29-88. [PMID: 26811286 DOI: 10.1016/bs.ircmb.2015.10.001] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Phosphatidylethanolamine (PE) is the second most abundant glycerophospholipid in eukaryotic cells. The existence of four only partially redundant biochemical pathways that produce PE, highlights the importance of this essential phospholipid. The CDP-ethanolamine and phosphatidylserine decarboxylase pathways occur in different subcellular compartments and are the main sources of PE in cells. Mammalian development fails upon ablation of either pathway. Once made, PE has diverse cellular functions that include serving as a precursor for phosphatidylcholine and a substrate for important posttranslational modifications, influencing membrane topology, and promoting cell and organelle membrane fusion, oxidative phosphorylation, mitochondrial biogenesis, and autophagy. The importance of PE metabolism in mammalian health has recently emerged following its association with Alzheimer's disease, Parkinson's disease, nonalcoholic liver disease, and the virulence of certain pathogenic organisms.
Collapse
Affiliation(s)
- Elizabeth Calzada
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ouma Onguka
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Khaire AA, Kale AA, Joshi SR. Maternal omega-3 fatty acids and micronutrients modulate fetal lipid metabolism: A review. Prostaglandins Leukot Essent Fatty Acids 2015; 98:49-55. [PMID: 25958298 DOI: 10.1016/j.plefa.2015.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 12/16/2022]
Abstract
It is well established that alterations in the mother's diet or metabolism during pregnancy has long-term adverse effects on the lipid metabolism in the offspring. There is growing interest in the role of specific nutrients especially omega-3 fatty acids in the pathophysiology of lipid disorders. A series of studies carried out in humans and rodents in our department have consistently suggested a link between omega-3 fatty acids especially docosahexaenoic acid and micronutrients (vitamin B12 and folic acid) in the one carbon metabolic cycle and its effect on the fatty acid metabolism, hepatic transcription factors and DNA methylation patterns. However the association of maternal intake or metabolism of these nutrients with fetal lipid metabolism is relatively less explored. In this review, we provide insights into the role of maternal omega-3 fatty acids and vitamin B12 and their influence on fetal lipid metabolism through various mechanisms which influence phosphatidylethanolamine-N-methyltransferase activity, peroxisome proliferator activated receptor, adiponectin signaling pathway and epigenetic process like chromatin methylation. This will help understand the possible mechanisms involved in fetal lipid metabolism and may provide important clues for the prevention of lipid disorders in the offspring.
Collapse
Affiliation(s)
- Amrita A Khaire
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune 411043, India
| | - Anvita A Kale
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune 411043, India
| | - Sadhana R Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune 411043, India.
| |
Collapse
|
49
|
Shumar SA, Fagone P, Alfonso-Pecchio A, Gray JT, Rehg JE, Jackowski S, Leonardi R. Induction of Neuron-Specific Degradation of Coenzyme A Models Pantothenate Kinase-Associated Neurodegeneration by Reducing Motor Coordination in Mice. PLoS One 2015; 10:e0130013. [PMID: 26052948 PMCID: PMC4460045 DOI: 10.1371/journal.pone.0130013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/15/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Pantothenate kinase-associated neurodegeneration, PKAN, is an inherited disorder characterized by progressive impairment in motor coordination and caused by mutations in PANK2, a human gene that encodes one of four pantothenate kinase (PanK) isoforms. PanK initiates the synthesis of coenzyme A (CoA), an essential cofactor that plays a key role in energy metabolism and lipid synthesis. Most of the mutations in PANK2 reduce or abolish the activity of the enzyme. This evidence has led to the hypothesis that lower CoA might be the underlying cause of the neurodegeneration in PKAN patients; however, no mouse model of the disease is currently available to investigate the connection between neuronal CoA levels and neurodegeneration. Indeed, genetic and/or dietary manipulations aimed at reducing whole-body CoA synthesis have not produced a desirable PKAN model, and this has greatly hindered the discovery of a treatment for the disease. OBJECTIVE, METHODS, RESULTS AND CONCLUSIONS Cellular CoA levels are tightly regulated by a balance between synthesis and degradation. CoA degradation is catalyzed by two peroxisomal nudix hydrolases, Nudt7 and Nudt19. In this study we sought to reduce neuronal CoA in mice through the alternative approach of increasing Nudt7-mediated CoA degradation. This was achieved by combining the use of an adeno-associated virus-based expression system with the synapsin (Syn) promoter. We show that mice with neuronal overexpression of a cytosolic version of Nudt7 (scAAV9-Syn-Nudt7cyt) exhibit a significant decrease in brain CoA levels in conjunction with a reduction in motor coordination. These results strongly support the existence of a link between CoA levels and neuronal function and show that scAAV9-Syn-Nudt7cyt mice can be used to model PKAN.
Collapse
Affiliation(s)
- Stephanie A. Shumar
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Paolo Fagone
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Adolfo Alfonso-Pecchio
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - John T. Gray
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Suzanne Jackowski
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Roberta Leonardi
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
50
|
Male-Specific Cardiac Dysfunction in CTP:Phosphoethanolamine Cytidylyltransferase (Pcyt2)-Deficient Mice. Mol Cell Biol 2015; 35:2641-57. [PMID: 25986609 DOI: 10.1128/mcb.00380-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/14/2015] [Indexed: 12/15/2022] Open
Abstract
Phosphatidylethanolamine (PE) is the most abundant inner membrane phospholipid. PE synthesis from ethanolamine and diacylglycerol is regulated primarily by CTP:phosphoethanolamine cytidylyltransferase (Pcyt2). Pcyt2(+/-) mice have reduced PE synthesis and, as a consequence, perturbed glucose and fatty acid metabolism, which gradually leads to the development of hyperlipidemia, obesity, and insulin resistance. Glucose and fatty acid uptake and the corresponding transporters Glut4 and Cd36 are similarly impaired in male and female Pcyt2(+/-) hearts. These mice also have similarly reduced phosphatidylinositol 3-kinase (PI3K)/Akt1 signaling and increased reactive oxygen species (ROS) production in the heart. However, only Pcyt2(+/-) males develop hypertension and cardiac hypertrophy. Pcyt2(+/-) males have upregulated heart AceI expression, heart phospholipids enriched in arachidonic acid and other n-6 polyunsaturated fatty acids, and dramatically increased ROS production in the aorta. In contrast, Pcyt2(+/-) females have unmodified heart phospholipids but have reduced heart triglyceride levels and altered expression of the structural genes Acta (low) and Myh7 (high). These changes together protect Pcyt2(+/-) females from cardiac dysfunction under conditions of reduced glucose and fatty acid uptake and heart insulin resistance. Our data identify Pcyt2 and membrane PE biogenesis as important determinants of gender-specific differences in cardiac lipids and heart function.
Collapse
|