1
|
Silva JG, Tavares L, Belew GD, Rodrigues JA, Araújo R, Gil AM, Jones JG. Impact of High-Fat Diet-induced Metabolic Dysfunction-associated Steatotic Liver Disease on Heart, Kidney, and Skeletal Muscle Metabolomes in Wild-Type Mice. J Proteome Res 2025; 24:2491-2504. [PMID: 40222045 DOI: 10.1021/acs.jproteome.5c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) can be recapitulated in mice fed a high-fat diet. The development of MASLD and the diet per se can both perturb metabolism in key extrahepatic tissues such as the heart, kidney, and skeletal muscle. To date, these alterations have not been well described in this animal model of diet-induced MASLD. Methodology: Male C57BL/6J mice were fed either standard (SC, n = 12) or high-fat chow (HF, n = 11) for 18 weeks. Metabolites were extracted from the heart, kidney, and skeletal muscle and analyzed by 1H nuclear magnetic resonance (NMR) spectroscopy, along with multivariate and univariate statistical analyses. Results: Kidney metabolite profiles exhibited the largest differences between HF and SC diets, followed by those of skeletal muscle and then the heart. Some alterations were common across all tissues, namely decreased trimethylamine and elevated levels of linoleic acid and polyunsaturated fatty acids in HF compared to SC (p < 0.05 for all three metabolites). Overall, the metabolite variations were consistent with shifts in carbohydrate and lipid substrate selection for oxidation, increased tissue stress in the heart and kidneys, and altered choline metabolism. These findings may serve as additional important descriptors of MASLD onset and progression.
Collapse
Affiliation(s)
- João G Silva
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Institute for Interdisciplinary Research (III-UC), Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra 3030-789, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ludgero Tavares
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
| | - Getachew D Belew
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, United States
| | - João A Rodrigues
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Rita Araújo
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - John G Jones
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
| |
Collapse
|
2
|
Liu X, Lu J, Ni X, He Y, Wang J, Deng Z, Zhang G, Shi T, Chen W. FASN promotes lipid metabolism and progression in colorectal cancer via the SP1/PLA2G4B axis. Cell Death Discov 2025; 11:122. [PMID: 40148316 PMCID: PMC11950308 DOI: 10.1038/s41420-025-02409-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Abnormal metabolic reprogramming is essential for tumorigenesis, metastasis, and the regulation of immune responses. Fatty acid synthase (FASN), a key enzyme in lipid metabolism, plays a crucial role in these processes. However, the relationship between FASN-mediated lipid reprogramming and the immune response in colorectal cancer (CRC) remains unclear. The present study demonstrated that FASN expression is elevated in CRC tissues and is significantly associated with poor prognosis. Functional experiments revealed that FASN promotes proliferation, migration, invasion, and phosphatidylcholine (PC) production in CRC cells. Additionally, in vivo experiments revealed that FASN knockdown significantly inhibits tumor growth and the spread of CRC cells to the lungs. Mechanistically, FASN, which is upregulated in CRC tissues, drives cancer cell proliferation, metastasis, and PC metabolism through the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of natural killer (NK) cells in a PC-dependent manner. These findings provide new insights into lipid metabolism and the immunobiology of CRC, suggesting potential targets for the treatment and prevention of CRC. Schematic diagram showing the mechanism by which FASN promotes cancer cell proliferation, metastasis, and PC metabolism in CRC via the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of NK cells in a PC-dependent manner. FFA free fatty acid, LPA lysophosphatidic acid, PA phosphatidate, DAG diglyceride, PC phosphatidylcholine, LPC lysophosphatidylcholine, CE cholesterol ester, TAG triacylglycerol.
Collapse
Affiliation(s)
- Xin Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiachun Lu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Xiangyu Ni
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Yuxin He
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Zilin Deng
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Yang J, Fan Y, Kang F, Yang Y, Wang Y, Liu Y, Han L. Phosphatidylcholine Cytidine Transferase α (CCTα) Affects LD Formation Through Fusion and Lipophagy in Bovine Mammary Epithelial Cells. Int J Mol Sci 2025; 26:2135. [PMID: 40076755 PMCID: PMC11901133 DOI: 10.3390/ijms26052135] [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: 12/28/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Phosphatidylcholine cytidine transferase α (CCTα) is a key rate-limiting enzyme in the CDP-choline pathway, the primary pathway for phosphatidylcholine (PC) synthesis in mammals. This study investigated the role of CCTα in lipid droplet (LD) formation, phospholipid synthesis, LD fusion, and lipophagy in bovine mammary epithelial cells (BMECs) through CCTα gene knockout (CCT-KO) and overexpression (CCT-OE). CCTα mRNA expression was significantly increased in bovine mammary gland tissue after lactation. In BMECs, CCTα was transferred from the nucleus to the endoplasmic reticulum and localized on LD surfaces in the presence of linoleic acid. Compared with normal BMECs (NC), CCTα knockout (CCT-KO) cells had significantly greater LD diameters (1.53 μm vs. 1.68 μm, p < 0.05), lower proportions of small LDs (<1 µm; 11.39% vs. 5.42%), and higher proportions of large LDs (>3 µm; 0.67% vs. 2.88%). In contrast, CCTα overexpression (CCT-OE) decreased the diameter of LDs to 1.18 μm (p < 0.01), increased the proportion of small LDs to 35.48%, and decreased the proportion of large LDs to 0.24%. CCTα knockout significantly decreased the PC content and the ratio of PC to PE, whereas CCTα overexpression increased the PC content and the ratio of PC to phosphatidyl ethanolamine (PE) (p < 0.05). The lipidomics analysis indicated that PC synthesis was significantly influenced by CCTα gene expression. Live cell observations showed that CCTα knockout promoted the fusion of small LDs into large LDs. In cells with CCT α overexpression, the expression of the microtubule-associated protein 1 light chain 3 (LC3) protein and the number of lysosomes was elevated, and the lysosomal phagocytosis of LDs was observed through transmission electron microscopy, thus indicating that CCTα overexpression enhanced lipophagy. In conclusion, these results suggest that CCTα plays a role in regulating LD formation by influencing PC synthesis, LD fusion, and lipophagy in BMECs.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Liu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (J.Y.); (Y.F.); (F.K.); (Y.Y.); (Y.W.)
| | - Liqiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; (J.Y.); (Y.F.); (F.K.); (Y.Y.); (Y.W.)
| |
Collapse
|
4
|
Pai AA, Chakraborty K, Dhara S, Raj A, Kariyil BJ, R A. Anti-dyslipidemic potential of sulfated glycosaminoglycan from rock oyster Saccostrea cucullata: An in vivo study. Chem Biol Interact 2025; 408:111371. [PMID: 39778732 DOI: 10.1016/j.cbi.2025.111371] [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: 06/23/2024] [Revised: 12/27/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
The rock oyster, Saccostrea cucullata, native to the Indo-Pacific region, is widely recognized for its nutritional and therapeutic benefits. A sulfated glycosaminoglycan (SCP-2) with β-(1 → 3)-GlcNSp and α-(1 → 4)-GlcAp as recurring units was isolated from S. cucullata. SCP-2 exhibited substantial 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) inhibition potential (IC50 0.65 mg/mL) in comparison with atorvastatin (IC50 0.72 mg/mL). An in vitro study of SCP-2 (0.1-160 μg/dL) revealed a 77-89 % reduction in triglyceride levels in Caco-2 cells after 4 days of incubation, similar to atorvastatin-treated cells (90 %). The efficacy of SCP-2 (at 90 mg/kg body weight) showed timely alleviation of triglyceride and cholesterol levels in tyloxapol-induced dyslipidemic rats (∼43 % and 81 % inhibition at 5 h), which was analogous to the atorvastatin treatment group (∼66 % and 71 %). Furthermore, SCP-2 (at 90 mg/kg body weight) showed mitigation in triglyceride (>50 %) and cholesterol levels (>25 %) in high-fat high-cholesterol (HFHC) diet-induced rats, similar to the lovastatin treatment group (approximately 62 % and 33 % inhibition on the 45th day). Histopathological studies of SCP-2 also showed recovery in ballooning degeneration, steatosis, and inflammation in liver tissues. Structure-activity relationship analysis suggested the importance of sulfate group in SCP-2 in contributing to its anti-dyslipidemic efficacy. The capability of SCP-2 to mitigate cholesterol, triglyceride, and HMGCR levels positions it as a promising functional food against dyslipidemia-related disorders.
Collapse
Affiliation(s)
- Ashwin Ashok Pai
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri, 574199, Karnataka State, India
| | - Kajal Chakraborty
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, 682018, Kerala State, India.
| | - Shubhajit Dhara
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri, 574199, Karnataka State, India
| | - Archana Raj
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Kerala Veterinary and Animal Sciences University, 680651, Thrissur, Kerala State, India
| | - Bibu John Kariyil
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary and Animal Sciences, Mannuthy, Kerala Veterinary and Animal Sciences University, 680651, Thrissur, Kerala State, India
| | - Anoopraj R
- Department of Veterinary Pathology, College of Veterinary and Animal Sciences, Mannuthy, Kerala Veterinary and Animal Sciences University, 680651, Thrissur, India
| |
Collapse
|
5
|
Wang L, Cheng C, Yu X, Guo L, Wan X, Xu J, Xiang X, Yang J, Kang J, Deng Q. Conversion of α-linolenic acid into n-3 long-chain polyunsaturated fatty acids: bioavailability and dietary regulation. Crit Rev Food Sci Nutr 2024:1-33. [PMID: 39686568 DOI: 10.1080/10408398.2024.2442064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
N-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs) are essential for physiological requirements and disease prevention throughout life but are not adequately consumed worldwide. Dietary supplementation with plant-derived α-linolenic acid (ALA) has the potential to rebalance the fatty acid profile and enhance health benefits but faces challenges such as high β-oxidation consumption, low hepatic conversion efficiency, and high oxidative susceptibility under stress. This review focuses on the metabolic fate and potential regulatory targets of ALA-containing lipids in vivo, specifically the pathway from the gastrointestinal tract to the lymph, blood circulation, and liver. We propose a hypothesis that positively regulates the conversion of ALA into n-3 LCPUFAs based on the model of "fast" or "slow" absorption, transport, and hepatic metabolic fate. Furthermore, the potential effects of dietary nutrients on the metabolic conversion of ALA into n-3 LCPUFAs are discussed. The conversion of ALA is differentially regulated by structured lipids, phospholipids, other lipids, carbohydrates, specific proteins, amino acids, polyphenols, vitamins, and minerals. Future research should focus on designing a steady-state and precise delivery system for ALA, coupled with specific nutrients or phytochemicals, to effectively improve its metabolic conversion and ultimately achieve synergistic regulation of nutrition and health effects.
Collapse
Affiliation(s)
- Lei Wang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Chen Cheng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xiao Yu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan, China
| | - Liang Guo
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xia Wan
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jiqu Xu
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Xia Xiang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jing Yang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| | - Jingxuan Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qianchun Deng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, and Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, Hubei, China
| |
Collapse
|
6
|
Chen S, Inui S, Aisyah R, Nakashima R, Kawaguchi T, Hinomoto M, Nakagawa Y, Sakuma T, Sotomaru Y, Ohshima N, Kumrungsee T, Ohkubo T, Yamamoto T, Miura Y, Suzuki T, Yanaka N. Role of Gpcpd1 in intestinal alpha-glycerophosphocholine metabolism and trimethylamine N-oxide production. J Biol Chem 2024:107965. [PMID: 39510189 DOI: 10.1016/j.jbc.2024.107965] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024] Open
Abstract
Glycerophosphocholine (GPC) is an intracellular metabolite in phosphatidylcholine metabolism and has been studied for endogenous choline supply in cells. GPC, as a water-soluble supplement, has been expected to play a role in preventing brain disorders; however, recent studies have shown that intake of high levels of choline-containing compounds is related to trimethylamine N-oxide (TMAO) production in the liver, which is reportedly associated with the progression of atherosclerosis. In this study, we aimed to explore the mechanisms underlying the intestinal absorption and metabolism of GPC. Caco-2 cell monolayer experiments showed that exogenously added GPC was hydrolyzed to choline in the apical medium, and the resulting choline was transported into the Caco-2 cells and further to the basolateral medium. Subsequently, we focused on glycerophosphodiesterase 1 (Gpcpd1/GDE5), which hydrolyzes GPC to choline in vitro and is widely expressed in the gastrointestinal epithelium. Our results revealed that the Gpcpd1 protein was located not only in cells but also in the medium in which Caco-2 cells were cultured. Gpcpd1 siRNA decreased the GPC-hydrolyzing activity both inside Caco-2 cells and in conditioned medium, suggesting the involvement of Gpcpd1 in luminal GPC metabolism. Finally, we generated intestinal epithelial-specific Gpcpd1-deficient mice and found that Gpcpd1 deletion in intestinal epithelial cells affected GPC metabolism in intestinal tissues and partially abolished the increase in blood TMAO levels induced by GPC administration. These observations demonstrate that Gpcpd1 triggers choline production from GPC in the intestinal lumen and is a key endogenous enzyme that regulates TMAO levels following GPC supplementation.
Collapse
Affiliation(s)
- Siyi Chen
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Shiho Inui
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Rahmawati Aisyah
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Ryoko Nakashima
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Tatsuya Kawaguchi
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Minori Hinomoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Yoshiko Nakagawa
- Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto 860-0811, Japan
| | - Tetsushi Sakuma
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 734-8553, Japan
| | - Noriyasu Ohshima
- Graduate School of Medicine, Gunma University, Gunma 371-8511, Japan
| | - Thanutchaporn Kumrungsee
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Takeshi Ohkubo
- Sendai Shirayuri Women's College, Sendai 981-3107, Japan
| | - Takashi Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Yutaka Miura
- Tokyo University of Agriculture and Technology, Tokyo 183-8538, Japan
| | - Takuya Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan.
| |
Collapse
|
7
|
Jiang X, Wang H, Nie K, Gao Y, Chen S, Tang Y, Wang Z, Su H, Dong H. Targeting lipid droplets and lipid droplet-associated proteins: a new perspective on natural compounds against metabolic diseases. Chin Med 2024; 19:120. [PMID: 39232826 PMCID: PMC11373146 DOI: 10.1186/s13020-024-00988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Lipid droplet (LD) is a metabolically active organelle, which changes dynamically with the metabolic state and energy requirements of cells. Proteins that either insert into the LD phospholipid monolayer or are present in the cytoplasm, playing a crucial role in lipid homeostasis and signaling regulation, are known as LD-associated proteins. METHODS The keywords "lipid droplets" and "metabolic diseases" were used to obtain literature on LD metabolism and pathological mechanism. After searching databases including Scopus, OVID, Web of Science, and PubMed from 2013 to 2024 using terms like "lipid droplets", "lipid droplet-associated proteins", "fatty liver disease", "diabetes", "diabetic kidney disease", "obesity", "atherosclerosis", "hyperlipidemia", "natural drug monomers" and "natural compounds", the most common natural compounds were identified in about 954 articles. Eventually, a total of 91 studies of 10 natural compounds reporting in vitro or in vivo studies were refined and summarized. RESULTS The most frequently used natural compounds include Berberine, Mangostin, Capsaicin, Caffeine, Genistein, Epigallocatechin-3-gallate, Chlorogenic acid, Betaine, Ginsenoside, Resveratrol. These natural compounds interact with LD-associated proteins and help ameliorate abnormal LDs in various metabolic diseases. CONCLUSION Natural compounds involved in the regulation of LDs and LD-associated proteins hold promise for treating metabolic diseases. Further research into these interactions may lead to new therapeutic applications.
Collapse
Affiliation(s)
- Xinyue Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueheng Tang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Su
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Arshad U, Zimpel R, Husnain A, Poindexter MB, Santos JEP. Effect of rumen-protected choline on fat digestibility and lymph metabolome in dairy cows. J Anim Physiol Anim Nutr (Berl) 2024; 108:950-964. [PMID: 38379267 DOI: 10.1111/jpn.13943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 12/08/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
Objectives were to determine the effects of supplementing rumen-protected choline (RPC) from an established source with low (L, 28.8%) or a prototype with less lipid coating protection and high (H, 60.0%) concentrations of choline chloride on digestibility of fat and supra-mammary lymph metabolome in feed-restricted cows. Pregnant, nonlactating Holstein cows (n = 33; 11/treatment) at mean (±standard deviation) 231 ± 4.7 days of gestation were blocked by body condition (4.23 ± 0.47) and assigned to receive 0 (CON) or 25.8 g/d of choline ion from L (L25.8) or H (H25.8). Cows were adapted to the diet and then fed-restricted to 42% of the net energy of lactation required for maintenance and pregnancy for 9 days. Intake of metabolizable methionine was maintained at 19 g/d. On Day 9, cows were fed 450 g of saturated fatty acids (SFA), and feces and blood were sampled continuously for 24 h. Supra-mammary lymph was sampled 6 h after feeding SFA and metabolome was characterized. Feeding RPC increased digestibility of fat (CON = 80.4 vs. RPC = 86.0 ± 1.9%) and reduced the concentration of haptoglobin in serum (CON = 174 vs. RPC = 77 ± 14 µg/ml) independent of source of RPC fed. Feeding RPC increased the concentrations of triacylglycerol in serum (CON = 15.1 vs. RPC = 17.8 ± 1.9 mg/dl) in feed-restricted cows after feeding SFA, and the increment tended to be greater for cows fed H25.8 than L25.8. Supplementing RPC tended to increase the concentrations of triacylglycerol (CON = 11.4 vs. RPC = 15.8 ± 3.4 mg/dl) in supra-mammary lymph. Feeding RPC increased the concentration of choline and affected the concentrations of analytes involved in metabolic pathways associated with amino acid metabolism and biosynthesis of phospholipids in lymph compared with CON. Feeding RPC, independent of source used, increased fat digestibility with some changes in lymph metabolome in cows under negative nutrient balance.
Collapse
Affiliation(s)
- Usman Arshad
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, USA
| | - Roney Zimpel
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, USA
| | - Ali Husnain
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, USA
| | - Michael B Poindexter
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, USA
| | - José E P Santos
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, USA
| |
Collapse
|
9
|
Ma Z, He Y, Li Y, Wang Q, Fang M, Yang Q, Gong Z, Xu L. Effects of Deoxynivalenol and Its Acetylated Derivatives on Lipid Metabolism in Human Normal Hepatocytes. Toxins (Basel) 2024; 16:294. [PMID: 39057934 PMCID: PMC11281666 DOI: 10.3390/toxins16070294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Deoxynivalenol (DON), 3-acetyldeoxynivalenol (3-ADON) and 15-acetyldeoxynivalenol (15-ADON) belong to type B trichothecenes that are widely detected in agricultural products as one of the most common classes of mycotoxins. In the present study, we aimed to characterize the alteration of lipid metabolism in normal human hepatocytes by poisoning with DON and its acetylated derivatives. After verifying the hepatotoxicity of the three toxins, DON, 15-ADON, and 3-ADON, the mRNA expression was determined by transcriptomics, and the results showed that DON and 15-ADON had a significant regulatory effect on the transcriptome, in which glycerophospholipid metabolism pathway and phospholipase D signaling pathways have not been reported in studies of DON and its acetylated derivatives. For further validation, we explored lipid metabolism in depth and found that PC (15:0/16:0), PC (16:1/18:3), PC (18:1/22:6), PC (16:0/16:0), PC (16:0/16:1), PC (16:1/18:1), PC (14:0/18:2), PE (14:0/16:0) and PE (18:1/18:3) were downregulated for all nine lipids. Combined with the transcriptome results, we found that hepatic steatosis induced by the three toxins, DON, 15-ADON and 3-ADON, was associated with altered expression of genes related to lipid oxidation, lipogenesis and lipolysis, and their effects on lipid metabolism in L-02 cells were mainly realized through the PC-PE cycle.
Collapse
Affiliation(s)
- Zhaoqing Ma
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuyun He
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuzhi Li
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-Derived Food for State Market Regulation, Wuhan 430075, China
| | - Qiao Wang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Min Fang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Qing Yang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Zhiyong Gong
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Lin Xu
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| |
Collapse
|
10
|
McPhee M, Lee J, Salsman J, Pinelli M, Di Cara F, Rosen K, Dellaire G, Ridgway ND. Nuclear lipid droplets in Caco2 cells originate from nascent precursors and in situ at the nuclear envelope. J Lipid Res 2024; 65:100540. [PMID: 38570093 PMCID: PMC11077042 DOI: 10.1016/j.jlr.2024.100540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
Intestinal epithelial cells convert excess fatty acids into triglyceride (TAG) for storage in cytoplasmic lipid droplets and secretion in chylomicrons. Nuclear lipid droplets (nLDs) are present in intestinal cells but their origin and relationship to cytoplasmic TAG synthesis and secretion is unknown. nLDs and related lipid-associated promyelocytic leukemia structures (LAPS) were abundant in oleate-treated Caco2 but less frequent in other human colorectal cancer cell lines and mouse intestinal organoids. nLDs and LAPS in undifferentiated oleate-treated Caco2 cells harbored the phosphatidate phosphatase Lipin1, its product diacylglycerol, and CTP:phosphocholine cytidylyltransferase (CCT)α. CCTα knockout Caco2 cells had fewer but larger nLDs, indicating a reliance on de novo PC synthesis for assembly. Differentiation of Caco2 cells caused large nLDs and LAPS to form regardless of oleate treatment or CCTα expression. nLDs and LAPS in Caco2 cells did not associate with apoCIII and apoAI and formed dependently of microsomal triglyceride transfer protein expression and activity, indicating they are not derived from endoplasmic reticulum luminal LDs precursors. Instead, undifferentiated Caco2 cells harbored a constitutive pool of nLDs and LAPS in proximity to the nuclear envelope that expanded in size and number with oleate treatment. Inhibition of TAG synthesis did affect the number of nascent nLDs and LAPS but prevented their association with promyelocytic leukemia protein, Lipin1α, and diacylglycerol, which instead accumulated on the nuclear membranes. Thus, nLD and LAPS biogenesis in Caco2 cells is not linked to lipoprotein secretion but involves biogenesis and/or expansion of nascent nLDs by de novo lipid synthesis.
Collapse
Affiliation(s)
- Michael McPhee
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jonghwa Lee
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jayme Salsman
- Depts of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marinella Pinelli
- Dept of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Francesca Di Cara
- Dept of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kirill Rosen
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Depts of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Neale D Ridgway
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
11
|
Guo Y, Liu F, Guo Y, Qu Y, Zhang Z, Yao J, Xu J, Li J. Untargeted Lipidomics Analysis Unravels the Different Metabolites in the Fat Body of Mated Bumblebee ( Bombus terrestris) Queens. Int J Mol Sci 2023; 24:15408. [PMID: 37895088 PMCID: PMC10607666 DOI: 10.3390/ijms242015408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
The fat body has important functions in energy, fertility, and immunity. In female insects, mating stimulates physiological, behavioral, and gene expression changes. However, it remains unclear whether the metabolites in the fat body are affected after the bumblebee (Bombus terrestris) queen mates. Here, the ultrastructure and lipid metabolites in fat body of mated queens were compared with those of virgins. The fat body weight of mated bumblebee queens was significantly increased, and the adipocytes were filled with lipid droplets. Using LC-MS/MS-based untargeted lipidomics, 949 and 748 differential metabolites were identified in the fat body of virgin and mated bumblebee queens, respectively, in positive and negative ion modes. Most lipid metabolites were decreased, especially some biomembrane components. In order to explore the relationship between the structures of lipid droplets and metabolite accumulation, transmission electron microscopy and fluorescence microscopy were used to observe the fat body ultrastructure. The size/area of lipid droplets was larger, and the fusion of lipid droplets was increased in the mated queen's fat body. These enlarged lipid droplets may store more energy and nutrients. The observed differences in lipid metabolites in the fat body of queens contribute to understanding the regulatory network of bumblebees post mating.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jilian Li
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.G.); (F.L.); (Y.G.); (Y.Q.); (Z.Z.); (J.Y.); (J.X.)
| |
Collapse
|
12
|
Lu X, Luo C, Wu J, Deng Y, Mu X, Zhang T, Yang X, Liu Q, Li Z, Tang S, Hu Y, Du Q, Xu J, Xie R. Ion channels and transporters regulate nutrient absorption in health and disease. J Cell Mol Med 2023; 27:2631-2642. [PMID: 37638698 PMCID: PMC10494301 DOI: 10.1111/jcmm.17853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 08/29/2023] Open
Abstract
Ion channels and transporters are ubiquitously expressed on cell membrane, which involve in a plethora of physiological process such as contraction, neurotransmission, secretion and so on. Ion channels and transporters is of great importance to maintaining membrane potential homeostasis, which is essential to absorption of nutrients in gastrointestinal tract. Most of nutrients are electrogenic and require ion channels and transporters to absorb. This review summarizes the latest research on the role of ion channels and transporters in regulating nutrient uptake such as K+ channels, Ca2+ channels and ion exchangers. Revealing the mechanism of ion channels and transporters associated with nutrient uptake will be helpful to provide new methods to diagnosis and find potential targets for diseases like diabetes, inflammatory bowel diseases, etc. Even though some of study still remain ambiguous and in early stage, we believe that ion channels and transporters will be novel therapeutic targets in the future.
Collapse
Affiliation(s)
- Xianmin Lu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Chen Luo
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jiangbo Wu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Ya Deng
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Xingyi Mu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Ting Zhang
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Xiaoxu Yang
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Qi Liu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Zhuo Li
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Siqi Tang
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Yanxia Hu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Qian Du
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jingyu Xu
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Rui Xie
- Department of GastroenterologyDigestive Disease Hospital, Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
13
|
Kosmerl E, Martínez-Sánchez V, Calvo MV, Jiménez-Flores R, Fontecha J, Pérez-Gálvez A. Food matrix impacts bioaccessibility and assimilation of acid whey-derived milk fat globule membrane lipids in Caco-2 cells. Front Nutr 2023; 10:1177152. [PMID: 37229475 PMCID: PMC10203207 DOI: 10.3389/fnut.2023.1177152] [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: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
The milk fat globule membrane (MFGM) imparts human health benefits ranging from improved immune system, gut, and brain function to improved cardiometabolic health. The industry's growing interest in introducing MFGM-enriched foods requires scientific evidence that the benefits derived from this compound are not affected by the formulation or processes that may alter its function, such as the digestion process. In this study, the impact of food matrices and supplementation levels on the bioaccessibility and assimilation of MFGM lipids in cell culture was investigated. Three food matrices including a protein-rich jelly, carbohydrate-rich cookie, and a carbohydrate- and fat-rich cookie with sunflower oil (SF-cookie) were supplemented with an MFGM ingredient derived from cottage cheese acid whey at 2, 5, and 10% (w/w). Each formulation underwent simulated digestion consisting of oral, gastric, and intestinal phases, and the micellar fraction was collected for both analysis and lipid assimilation in Caco-2 intestinal cells. The micellar fractions were diluted and applied to the cells for 4 h. A lipidomic approach was used to assess the lipid profiles of micellar fractions and intestinal cells. The micelles from digested jellies, cookies, and SF-cookies containing MFGM showed a distinct separation using partial least squares discriminant analysis (PLS-DA). Both correlation loadings and variable importance in projection (VIP) scores demonstrated a tendency of MFGM polar lipids (ceramides, glucosylceramides) for micelles from digested jelly, whereas micelles from digested cookies were associated with MFGM neutral lipids (free fatty acids, cholesterol, etc.). The effect of supplementation level on the micellar lipid profiles reinforced this pattern. The lipid profiles of intestinal cells after incubation with the micellar fractions differed considerably from the corresponding micellar lipid profiles. Specifically, the SF-cookie-treated cells were associated with a greater abundance of PUFA relative to jelly- and cookie-treated cells; however, increasing MFGM supplementation showed irregular patterns and rearrangement of cellular lipid profiles, suggesting the cells' role in regulating lipid metabolism in response to nutritional stimuli. The nature of lipid micellarization and assimilation in intestinal cells from MFGM-containing food formulations echoes the complexity of lipids inherent to the MFGM itself, suggesting the need for application-based MFGM supplementation.
Collapse
Affiliation(s)
- Erica Kosmerl
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Victoria Martínez-Sánchez
- Group of Chemistry and Biochemistry of Pigments, Instituto de la Grasa (CSIC), Sevilla, Spain
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CSIC-UAM), Madrid, Spain
| | - María V. Calvo
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CSIC-UAM), Madrid, Spain
| | - Rafael Jiménez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Javier Fontecha
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CSIC-UAM), Madrid, Spain
| | - Antonio Pérez-Gálvez
- Group of Chemistry and Biochemistry of Pigments, Instituto de la Grasa (CSIC), Sevilla, Spain
| |
Collapse
|
14
|
Dorighello G, McPhee M, Halliday K, Dellaire G, Ridgway N. Differential contributions of phosphotransferases CEPT1 and CHPT1 to phosphatidylcholine homeostasis and lipid droplet biogenesis. J Biol Chem 2023; 299:104578. [PMID: 36871755 PMCID: PMC10166788 DOI: 10.1016/j.jbc.2023.104578] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The CDP-choline (Kennedy) pathway culminates with the synthesis of phosphatidylcholine (PC) and phosphatidylethanolamine (PE) by choline/ethanolamine phosphotransferase 1 (CEPT1) in the endoplasmic reticulum (ER), and PC synthesis by choline phosphotransferase 1 (CHPT1) in the Golgi apparatus. Whether the PC and PE synthesized by CEPT1 and CHPT1 in the ER and Golgi apparatus has different cellular functions has not been formally addressed. Here we used CRISPR editing to generate CEPT1-and CHPT1-knockout (KO) U2OS cells to assess the differential contribution of the enzymes to feed-back regulation of nuclear CTP:phosphocholine cytidylyltransferase (CCT)α, the rate-limiting enzyme in PC synthesis, and lipid droplet (LD) biogenesis. We found that CEPT1-KO cells had a 50% and 80% reduction in PC and PE synthesis, respectively, while PC synthesis in CHPT1-KO cells was also reduced by 50%. CEPT1 knockout caused the post-transcriptional induction of CCTα protein expression as well as its dephosphorylation and constitutive localization on the inner nuclear membrane and nucleoplasmic reticulum. This activated CCTα phenotype was prevented by incubating CEPT1-KO cells with PC liposomes to restore end-product inhibition. Additionally, we determined that CEPT1 was in close proximity to cytoplasmic LDs, and CEPT1 knockout resulted in the accumulation of small cytoplasmic LDs, as well as increased nuclear LDs enriched in CCTα. In contrast, CHPT1 knockout had no effect on CCTα regulation or LD biogenesis. Thus, CEPT1 and CHPT1 contribute equally to PC synthesis; however, only PC synthesized by CEPT1 in the ER regulates CCTα and the biogenesis of cytoplasmic and nuclear LDs.
Collapse
Affiliation(s)
- Gabriel Dorighello
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2
| | - Michael McPhee
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2
| | - Katie Halliday
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2
| | - Graham Dellaire
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2; Depts of Pathology and Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2
| | - NealeD Ridgway
- Depts of Pediatrics and Biochemistry & Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada B3H4R2.
| |
Collapse
|
15
|
Bai ZY, Zheng H, Luo Z, Hogstrand C, Wang LJ, Song YF. Dietary Choline Mitigates High-Fat Diet-Impaired Chylomicrons Assembly via UPRer Modulated by perk DNA Methylation. Cells 2022; 11:cells11233848. [PMID: 36497107 PMCID: PMC9741040 DOI: 10.3390/cells11233848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
High-fat diets (HFD) lead to impairment of chylomicrons (CMs) assembly and adversely influence intestinal lipid homeostasis. However, the mechanisms of HFD impairing CMs assembly have yet to be fully understood. Additionally, although choline, as a lipid-lowering agent, has been widely used and its deficiency has been closely linked to non-alcoholic steatohepatitis (NASH), the contribution of choline by functioning as a methyl donor in alleviating HFD-induced intestinal lipid deposition is unknown. Thus, this study was conducted to determine the mechanism of HFD impairing CMs assembly and also tested the effect of choline acting as a methyl donor in this process. To this end, in this study, four diets (control, HFD, choline and HFD + choline diet) were fed to yellow catfish for 10 weeks in vivo and their intestinal epithelial cells were isolated and incubated for 36 h in fatty acids (FA) with or without choline solution combining si-perk transfection in vitro. The key findings from this study as follows: (1) HFD caused impairment of CMs assembly main by unfolded protein response (UPRer). HFD activated perk and then induced UPRer, which led to endoplasmic reticulum dysfunction and further impaired CMs assembly via protein-protein interactions between Perk and Apob48. (2) Choline inhibited the transcriptional expression level of perk via activating the -211 CpG methylation site, which initiated the subsequent ameliorating effect on HFD-impaired CMs assembly and intestinal lipid dysfunction. These results provide a new insight into direct crosstalk between UPRer and CMs assembly, and also emphasize the critical contribution of choline acting as a methyl donor and shed new light on choline-deficient diet-induced NASH.
Collapse
Affiliation(s)
- Zhen-Yu Bai
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Hua Zheng
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Christer Hogstrand
- Department of Nutritional Sciences, School of Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Ling-Jiao Wang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Yu-Feng Song
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: ; Tel.: +86-27-8728-2113; Fax: +86-27-8728-2114
| |
Collapse
|
16
|
Stone SJ. Mechanisms of intestinal triacylglycerol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159151. [PMID: 35296424 DOI: 10.1016/j.bbalip.2022.159151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
Abstract
Triacylglycerols are a major source of stored energy that are obtained either from the diet or can be synthesized to some extent by most tissues. Alterations in pathways of triacylglycerol metabolism can result in their excessive accumulation leading to obesity, insulin resistance, cardiovascular disease and nonalcoholic fatty liver disease. Most tissues in mammals synthesize triacylglycerols via the glycerol 3-phosphate pathway. However, in the small intestine the monoacylglycerol acyltransferase pathway is the predominant pathway for triacylglycerol biosynthesis where it participates in the absorption of dietary triacylglycerol. In this review, the enzymes that are part of both the glycerol 3-phosphate and monoacylglycerol acyltransferase pathways and their contributions to intestinal triacylglycerol metabolism are reviewed. The potential of some of the enzymes involved in triacylglycerol synthesis in the small intestine as possible therapeutic targets for treating metabolic disorders associated with elevated triacylglycerol is briefly discussed.
Collapse
Affiliation(s)
- Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
17
|
Zhang G, Tobolski D, Zwierzchowski G, Mandal R, Wishart DS, Ametaj BN. Identification of Serum-Predictive Biomarkers for Subclinical Mastitis in Dairy Cows and New Insights into the Pathobiology of the Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1724-1746. [PMID: 35098717 DOI: 10.1021/acs.jafc.1c07281] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Targeted direct injection/liquid chromatography coupled to tandem mass spectrometry-based metabolomics was employed to identify metabolite alterations that could differentiate subclinical mastitis (SCM) from control (CON) dairy cows at -8, -4, disease diagnosis, +4 and +8 wks relative to parturition. We identified and measured 128 metabolites in the serum. Univariate analysis revealed significant alterations of serum metabolites at all five time points studied. By applying multivariate analyses including principle component analysis and partial least squares-discriminant analysis, some of the metabolites were found to have the strongest power for discriminating the SCM from CON cows. The top five metabolites with the greatest variable importance in projection values were selected as potential biomarkers for SCM. A set of five serum metabolites including lysine, ornithine, isoleucine, LysoPC a C17:0, and leucine at -8 wks and five other metabolites including lysine, leucine, isoleucine, kynurenine, and sphingomyelin (SM) C26:0 at -4 wks prepartum were determined as predictive biomarkers for SCM, which provided highly predictive capabilities with AUC (area under the curve) at 1.00. Five metabolites including lysine, leucine, isoleucine, kynurenine, and SM C26:1 in the serum were identified as diagnostic biomarkers for SCM with the AUC of 1.00. Moreover, we observed that distinct metabolic pathways were affected in SCM cows including lysine degradation, biotin, cysteine, methionine, and glutathione metabolism, valine, leucine, and isoleucine biosynthesis and degradation, and aminoacyl-tRNA biosynthesis prior to and during the occurrence of the disease. Results of this study showed that metabolomics analyses can be used to identify susceptible cows to SCM starting from -8 and -4 wks prepartum and that blood can be used to diagnose cows with SCM.
Collapse
Affiliation(s)
- Guanshi Zhang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton AB T6G 2P5, Canada
| | - Dawid Tobolski
- Department of Internal Diseases with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, 14 Oczapowskiego Str., Olsztyn 10-718, Poland
| | - Grzegorz Zwierzchowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton AB T6G 2P5, Canada
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, 1a Oczapowskiego Str., Olsztyn 10-719, Poland
| | - Rupasri Mandal
- Departments of Biological and Computer Sciences, University of Alberta, Edmonton AB T6G 2P5, Canada
| | - David S Wishart
- Departments of Biological and Computer Sciences, University of Alberta, Edmonton AB T6G 2P5, Canada
| | - Burim N Ametaj
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton AB T6G 2P5, Canada
| |
Collapse
|
18
|
Zembroski AS, Xiao C, Buhman KK. The Roles of Cytoplasmic Lipid Droplets in Modulating Intestinal Uptake of Dietary Fat. Annu Rev Nutr 2021; 41:79-104. [PMID: 34283920 DOI: 10.1146/annurev-nutr-110320-013657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dietary fat absorption is required for health but also contributes to hyperlipidemia and metabolic disease when dysregulated. One step in the process of dietary fat absorption is the formation of cytoplasmic lipid droplets (CLDs) in small intestinal enterocytes; these CLDs serve as dynamic triacylglycerol storage organelles that influence the rate at which dietary fat is absorbed. Recent studies have uncovered novel factors regulating enterocyte CLD metabolism that in turn influence the absorption of dietary fat. These include peroxisome proliferator-activated receptor α activation, compartmentalization of different lipid pools, the gut microbiome, liver X receptor and farnesoid X receptor activation, obesity, and physiological factors stimulating CLD mobilization. Understanding how enterocyte CLD metabolism is regulated is key in modulating the absorption of dietary fat in the prevention of hyperlipidemia and its associated metabolic disorders. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Alyssa S Zembroski
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907, USA;
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907, USA;
| |
Collapse
|
19
|
Yu J, Wu C, Wu Q, Huang J, Fu W, Xie X, Li W, Tang W, Xu C, Jin G. PCYT1A suppresses proliferation and migration via inhibiting mTORC1 pathway in lung adenocarcinoma. Biochem Biophys Res Commun 2020; 529:353-361. [PMID: 32703435 DOI: 10.1016/j.bbrc.2020.05.164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 12/26/2022]
Abstract
Lung cancer is one of most common malignant cancer worldwide. It is emerging that PCYT1A, a rate-limiting enzyme required for the biosynthesis of phosphatidylcholine, is associated with cancer progression. However, the biological functions and underlying molecular mechanisms of PCYT1A in lung adenocarcinoma is still unknown. Here we found that PCYT1A suppressed lung adenocarcinoma cancer cell proliferation and migration. Mechanically, PCYT1A served as a novel negative regulator of mTORC1 signaling. PCYT1A knockdown enhanced the malignant proliferation and migration of lung adenocarcinoma cells by activating mTORC1. The promoting effects of PCYT1A silencing on cell proliferation and migration could be abolished when mTORC1 signaling was inhibited by rapamycin or RAPTOR depletion. Importantly, PCYT1A high expression predicted longer survival of lung cancer patients. The expression of PCYT1A was also negatively correlated with mTORC1 activation in the clinical lung cancer samples. We therefore reveal that PCYT1A suppresses proliferation and migration by inhibiting the mTORC1 signaling pathway in lung adenocarcinoma. PCYT1A shows as a potential promising biomarker in lung adenocarcinoma.
Collapse
Affiliation(s)
- Jing Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Changtao Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China; Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Qi Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jiafeng Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wenjuan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xuemei Xie
- Department of Pathology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637100, China
| | - Wen Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine University of Electronic Science and Technology of China, Chengdu, 610047, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Chuan Xu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine University of Electronic Science and Technology of China, Chengdu, 610047, China.
| | - Guoxiang Jin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), And Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
20
|
Yue L, McPhee MJ, Gonzalez K, Charman M, Lee J, Thompson J, Winkler DFH, Cornell RB, Pelech S, Ridgway ND. Differential dephosphorylation of CTP:phosphocholine cytidylyltransferase upon translocation to nuclear membranes and lipid droplets. Mol Biol Cell 2020; 31:1047-1059. [PMID: 32186954 PMCID: PMC7346725 DOI: 10.1091/mbc.e20-01-0014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CTP:phosphocholine cytidylyltransferase-alpha (CCTα) and CCTβ catalyze the rate-limiting step in phosphatidylcholine (PC) biosynthesis. CCTα is activated by association of its α-helical M-domain with nuclear membranes, which is negatively regulated by phosphorylation of the adjacent P-domain. To understand how phosphorylation regulates CCT activity, we developed phosphosite-specific antibodies for pS319 and pY359+pS362 at the N- and C-termini of the P-domain, respectively. Oleate treatment of cultured cells triggered CCTα translocation to the nuclear envelope (NE) and nuclear lipid droplets (nLDs) and rapid dephosphorylation of pS319. Removal of oleate led to dissociation of CCTα from the NE and increased phosphorylation of S319. Choline depletion of cells also caused CCTα translocation to the NE and S319 dephosphorylation. In contrast, Y359 and S362 were constitutively phosphorylated during oleate addition and removal, and CCTα-pY359+pS362 translocated to the NE and nLDs of oleate-treated cells. Mutagenesis revealed that phosphorylation of S319 is regulated independently of Y359+S362, and that CCTα-S315D+S319D was defective in localization to the NE. We conclude that the P-domain undergoes negative charge polarization due to dephosphorylation of S319 and possibly other proline-directed sites and retention of Y359 and S362 phosphorylation, and that dephosphorylation of S319 and S315 is involved in CCTα recruitment to nuclear membranes.
Collapse
Affiliation(s)
- Lambert Yue
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Michael J McPhee
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kevin Gonzalez
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Mark Charman
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jonghwa Lee
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jordan Thompson
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Dirk F H Winkler
- Kinexus Bioinformatics Corporation, Vancouver, BC V6P 6T3, Canada
| | - Rosemary B Cornell
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Steven Pelech
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC V6T 2B5, Canada.,Kinexus Bioinformatics Corporation, Vancouver, BC V6P 6T3, Canada
| | - Neale D Ridgway
- Department of Pediatrics and Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
21
|
Guttmann S, Nadzemova O, Grünewald I, Lenders M, Brand E, Zibert A, Schmidt HH. ATP7B knockout disturbs copper and lipid metabolism in Caco-2 cells. PLoS One 2020; 15:e0230025. [PMID: 32155648 PMCID: PMC7064347 DOI: 10.1371/journal.pone.0230025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/18/2020] [Indexed: 12/30/2022] Open
Abstract
Intestinal cells control delivery of lipids to the body by adsorption, storage and secretion. Copper (Cu) is an important trace element and has been shown to modulate lipid metabolism. Mutation of the liver Cu exporter ATP7B is the cause of Wilson disease and is associated with Cu accumulation in different tissues. To determine the relationship of Cu and lipid homeostasis in intestinal cells, a CRISPR/Cas9 knockout of ATP7B (KO) was introduced in Caco-2 cells. KO cells showed increased sensitivity to Cu, elevated intracellular Cu storage, and induction of genes regulating oxidative stress. Chylomicron structural protein ApoB48 was significantly downregulated in KO cells by Cu. Apolipoproteins ApoA1, ApoC3 and ApoE were constitutively induced by loss of ATP7B. Formation of small sized lipid droplets (LDs) was enhanced by Cu, whereas large sized LDs were reduced. Cu reduced triglyceride (TG) storage and secretion. Exposure of KO cells to oleic acid (OA) resulted in enhanced TG storage. The findings suggest that Cu represses intestinal TG lipogenesis, while loss of ATP7B results in OA-induced TG storage.
Collapse
Affiliation(s)
- Sarah Guttmann
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Oksana Nadzemova
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Inga Grünewald
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Münster, Germany
| | - Malte Lenders
- Department of Nephrology, Hypertension and Rheumatology, Internal Medicine D, University Hospital Muenster, Münster, Germany
| | - Eva Brand
- Department of Nephrology, Hypertension and Rheumatology, Internal Medicine D, University Hospital Muenster, Münster, Germany
| | - Andree Zibert
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Hartmut H. Schmidt
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
22
|
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
|
23
|
D'Aquila T, Zembroski AS, Buhman KK. Diet Induced Obesity Alters Intestinal Cytoplasmic Lipid Droplet Morphology and Proteome in the Postprandial Response to Dietary Fat. Front Physiol 2019; 10:180. [PMID: 30890954 PMCID: PMC6413465 DOI: 10.3389/fphys.2019.00180] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/13/2019] [Indexed: 12/31/2022] Open
Abstract
Dietary fat absorption by the small intestine is an efficient, multistep process that regulates the uptake and delivery of essential nutrients and energy. Fatty acids taken up by enterocytes, the absorptive cells of the small intestine, are resynthesized into triacylglycerol (TAG) and either secreted in chylomicrons or temporarily stored in cytoplasmic lipid droplets (CLDs). Proteins that associate with CLDs are thought to regulate the dynamics of TAG storage and mobilization. It is currently unclear what effect diet induced obesity (DIO) has on the balance between dietary fat storage and secretion. Specifically, there is limited knowledge of how DIO affects the level and diversity of proteins that associate with CLDs and regulate CLD dynamics. In the current study, we characterize CLDs from lean and DIO mice through histological and proteomic analyses. We demonstrate that DIO mice have larger intestinal CLDs compared to lean mice in response to dietary fat. Additionally, we identified 375 proteins in the CLD fraction isolated from enterocytes of lean and DIO mice. We identified a subgroup of lipid related proteins that are either increased or unique to the DIO CLD proteome. These proteins are involved in steroid synthesis, TAG synthesis, and lipolysis. This analysis expands our knowledge of the effect of DIO on the process of dietary fat absorption in the small intestine (D’Aquila, 2016).
Collapse
Affiliation(s)
- Theresa D'Aquila
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Alyssa S Zembroski
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| |
Collapse
|