1
|
Zhao C, Wang S, Li B, Zhang Z, Yu W, Yu H. Hepatic lipidomics analysis reveals the anti-obesity effects of insoluble dietary fiber from okara combined with intermittent fasting treatment in high-fat diet-fed mice. Front Nutr 2025; 12:1549105. [PMID: 40336964 PMCID: PMC12055546 DOI: 10.3389/fnut.2025.1549105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Emerging evidence has revealed that supplementation with insoluble dietary fiber (IDF) improves lipid metabolic disorders caused by a high-fat diet (HFD). Except for dietary supplementation, intermittent fasting (IF) has received widespread attention as a new dietary strategy against obesity. We hypothesized that IDF combined with IF treatment may synergistically alleviate HFD-induced lipid metabolic imbalance. Methods This study integrated biochemical analysis with hepatic lipidomics to explore lipid biomarkers and potential mechanisms associated with the anti-obesity effects of IDF combined with IF treatment. Results and discussion The results indicated that IDF combined with IF treatment improved metabolic parameters associated with the obesity phenotype. Lipidomics analysis revealed that IDF combined with IF treatment altered hepatic lipid metabolic patterns in HFD-fed mice. Moreover, 15 differentially regulated lipid species were identified as lipid biomarkers. Moreover, the expression of lipogenesis-, lipid oxidation-, and cholesterol metabolism-related genes was also regulated. Our results indicate that IDF combined with IF treatment ameliorates lipid metabolism disorders in HFD-fed mice by regulating hepatic lipid metabolites and related gene expression, providing evidence for its anti-obesity effects.
Collapse
Affiliation(s)
- Chenhao Zhao
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| | - Bo Li
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhao Zhang
- Shandong Sinoglory Health Food Co., Ltd., Liaocheng, China
| | - Wei Yu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, China
| |
Collapse
|
2
|
Sehgal R, Jähnert M, Lazaratos M, Speckmann T, Schumacher F, Kleuser B, Ouni M, Jonas W, Schürmann A. Altered liver lipidome markedly overlaps with human plasma lipids at diabetes risk and reveals adipose-liver interaction. J Lipid Res 2025; 66:100767. [PMID: 40044043 PMCID: PMC11997378 DOI: 10.1016/j.jlr.2025.100767] [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: 11/11/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Present study explores the role of liver lipidome in driving T2D-associated metabolic changes. Elevated liver triacylglycerols, reduced PUFAs, and 86 differentially abundant lipid species were identified in diabetes-prone mice. Of these altered lipid species, 82 markedly overlap with human plasma lipids associated with T2D/CVD risk. Pathway enrichment highlighted sphingolipid metabolism, however, only five of all genes involved in the pathway were differentially expressed in the liver. Interestingly, overlap with adipose tissue transcriptome was much higher (57 genes), pointing toward an active adipose-liver interaction. Next, the integration of liver lipidome and transcriptome identified strongly correlated lipid-gene networks highlighting ceramide [Cer(22:0)], dihydroceramide(24:1), and triacylglycerol(58:6) playing a central role in transcriptional regulation. Putative molecular targets of Cer(22:0) were altered (Cyp3a44, Tgf-β1) in primary mouse hepatocytes treated with Cer(22:0). Early alteration of liver lipidome markedly depends on adipose tissue expression pattern and provides substantial evidence linking early liver lipidome alterations and risk of T2D.
Collapse
Affiliation(s)
- Ratika Sehgal
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michail Lazaratos
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Thilo Speckmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Burkhard Kleuser
- Freie Universität Berlin, Institute of Pharmacy, Berlin, Germany
| | - Meriem Ouni
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Wenke Jonas
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
3
|
Rajendran R, Suman S, Divakaran SJ, Swatikrishna S, Tripathi P, Jain R, Sagar K, Rajakumari S. Sesaminol alters phospholipid metabolism and alleviates obesity-induced NAFLD. FASEB J 2024; 38:e23835. [PMID: 39037555 DOI: 10.1096/fj.202400412rr] [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: 02/23/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
The prevalence of obesity-induced non-alcoholic fatty liver disease (NAFLD) and insulin resistance is increasing worldwide. We previously demonstrated that sesaminol increases thermogenesis in adipocytes, improves insulin sensitivity, and mitigates obesity in mice. In this study, we demonstrated that sesaminol increased mitochondrial activity and reduced ROS production in hepatocytes. Therefore, we delve into the metabolic action of sesaminol in obesity-induced NAFLD or metabolic dysfunction-associated liver disease (MAFLD). Here, we report that sesaminol induces OXPHOS proteins and mitochondrial function in vivo. Further, our data suggest that sesaminol administration reduces hepatic triacylglycerol accumulation and LDL-C levels. Prominently, the lipidomics analyses revealed that sesaminol administration decreased the major phospholipids such as PC, PE, PI, CL, and PS to maintain membrane lipid homeostasis in the liver upon HFD challenge. Besides, SML reduced ePC and SM molecular species and increased PA levels in the HFD-fed mice. Also, sesaminol renders anti-inflammatory properties and dampens fibrosis markers in the liver. Remarkably, SML lowers the hepatic levels of ALT and AST enzymes and alleviates NAFLD in diet-induced obese mice. The molecular docking analysis identifies peroxisome proliferator-activated receptors as potential endogenous receptors for sesaminol. Together, our study demonstrates plant lignan sesaminol as a potential small molecule that alters the molecular species of major phospholipids, including sphingomyelin and ether-linked PCs in the liver tissue, improves metabolic parameters, and alleviates obesity-induced fatty liver disease in mice.
Collapse
Affiliation(s)
- Rajprabu Rajendran
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sanskriti Suman
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Soumya Jaya Divakaran
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sahu Swatikrishna
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Purnima Tripathi
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Rashi Jain
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Karan Sagar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sona Rajakumari
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
4
|
Xu J, Shi M, Chen L, Chi S, Zhang S, Cao J, Tan B, Xie S. Muscular lipidomics and transcriptomics reveal the effects of bile acids on lipid metabolism in high-fat diet-fed grouper. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:127-143. [PMID: 36826624 DOI: 10.1007/s10695-023-01176-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Little information is available on how exogenous bile acids alter lipid metabolism in muscle of fish. In the present study, an 8-week feeding trial were used to investigate the impacts of bile acids on lipid deposition, lipid metabolism, lipidomics, and transcriptomics in muscle of pearl gentian grouper (Epinephelus fuscoguttatus♀ × E. lanceolatus♂) fed a high-fat diet (HD). The HD treatment significantly increased the crude lipid content, while bile acids diet (BD) treatment decreased it (p = 0.057). BD treatment significantly decreased triglycerides level and significantly increased phosphatidylcholines, phosphatidylethanolamines, and phosphatidylglycerol levels. The contents of TG (17:0/18:2/18:2), TG (17:1/18:2/22:6), PC (6:0/22:1), PC (9:0/26:1), PC (26:1/6:0), PC (17:2/18:2), PE (16:0/18:1), PE (18:0/17:1), PG (18:0/20:5), PG (18:3/20:5), PG (19:0/16:1), and PG (18:0/18:1) in muscle were well response to dietary lipid level and bile acids supplementation. HD and BD groups induced a variety of adaptive metabolic responses in transcriptomics. HD treatment increased the lipogenesis and decreased lipolysis, whereas BD treatment decreased the lipogenesis and increased lipolysis. Present study revealed the improvement of muscular lipid metabolism and lipid composition in response to bile acids administration in pearl gentian grouper.
Collapse
Affiliation(s)
- Jia Xu
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning, China
| | - Menglin Shi
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Liutong Chen
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Shuyan Chi
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, China
| | - Shuang Zhang
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, China
| | - Junming Cao
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, China
| | - Beiping Tan
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China.
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China.
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, China.
| | - Shiwei Xie
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China.
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, China.
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, China.
| |
Collapse
|
5
|
Haroun N, Bennour I, Seipelt E, Astier J, Sani L, Tardivel C, Svilar L, Martin JC, Mounien L, Landrier JF. Maternal Vitamin D Deficiency in Mice Sex-Dependently Affects Hepatic Lipid Accumulation in Offspring. Mol Nutr Food Res 2024; 68:e2300290. [PMID: 38010607 DOI: 10.1002/mnfr.202300290] [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: 05/05/2023] [Revised: 10/18/2023] [Indexed: 11/29/2023]
Abstract
SCOPE Vitamin D deficiency (VDD) is becoming a global issue and low 25-hydroxyvitamin D (25(OH)D) plasma levels have been linked to hepatic steatosis in adulthood. Nevertheless, the impact of maternal VDD on lipid metabolism and hepatic steatosis remains poorly documented, especially under obesogenic condition. The goal of this study is to assess the effects of maternal VDD on hepatic lipid accumulation in adult offspring fed a normal or obesogenic diet. METHODS AND RESULTS Several approaches are implemented including histology and lipidomics on the liver in both males and females. No major impact of high-fat (HF) or VDD is observed at histological level in both males and females. Nevertheless, in males born from VDD mice and fed an HF diet, an increase of total lipids and modulation of the relative lipid species distribution characterized by a decrease of triglycerides and increase of phospholipids is observed. In female no major lipid profile is noticed. CONCLUSION Maternal VDD combined with a HF diet in male may predispose to hepatic hypertrophia, with a specific lipid profile. Such observations reinforce our knowledge of the impact of maternal VDD on hepatic programming in the offspring.
Collapse
Affiliation(s)
- Nicole Haroun
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
| | - Imene Bennour
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
| | - Eva Seipelt
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
| | - Julien Astier
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
| | - Léa Sani
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
| | - Catherine Tardivel
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
- Biomet, C2VN, CriBiom, Marseille, 13000, France
| | - Ljubica Svilar
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
- Biomet, C2VN, CriBiom, Marseille, 13000, France
| | - Jean-Charles Martin
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
- Biomet, C2VN, CriBiom, Marseille, 13000, France
| | - Lourdes Mounien
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
- PhenoMARS, C2VN, CriBiom, Marseille, 13000, France
| | - Jean François Landrier
- Aix-Marseille Université, C2VN, INRAE, INSERM, Marseille, 13000, France
- PhenoMARS, C2VN, CriBiom, Marseille, 13000, France
| |
Collapse
|
6
|
Fan X, Wang R, Song Y, Wang X, Liu Y, Wang X, Xu J, Xue C. Effects of high-sugar, high-cholesterol, and high-fat diet on phospholipid profile of mouse tissues with a focus on the mechanism of plasmalogen synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2023:159345. [PMID: 37268055 DOI: 10.1016/j.bbalip.2023.159345] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
High-sugar diet (HSD), high-cholesterol diet (HCD), and high-fat diet (HFD) all modulate the levels of lipids. However, there is a lack of comparative data on the effects of different diets on phospholipids (PLs). Given their important role in physiology and disease, there has been an increasing focus on altered PLs in liver and brain disorders. This study aims to determine the effects of HSD, HCD, and HFD for 14-week feeding on the PL profile of the mouse liver and hippocampus. Quantitative analysis of 116 and 113 PL molecular species in liver and hippocampus tissues revealed that the HSD, HCD, and HFD significantly affected the PLs in liver and hippocampus, especially decreased the levels of plasmenylethanolamine (pPE) and phosphatidylethanolamine (PE). Overall, the impact of HFD on liver PLs was more significant, consistent with the morphological changes in the liver. Compared to HSD and HCD, HFD induced a significant decrease in PC (P-16:0/18:1) and an increase in LPE (18:0) and LPE (18:1) in liver. In the liver of mice fed with different diets, the expression of the key enzymes Gnpat, Agps in the pPE biosynthesis pathway and peroxisome-associated membrane proteins pex14p were decreased. In addition, all diets significantly reduced the expression of Gnpat, pex7p, and pex16p in hippocampus tissue. In conclusion, HSD, HCD, and HFD enhanced lipid accumulation in the liver, led to liver injury, significantly affected the liver and hippocampus PLs, and decreased the expression of genes related to plasmalogen synthesis in mouse liver and hippocampus, which caused severe plasmalogen reduction.
Collapse
Affiliation(s)
- Xiaowei Fan
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Rui Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Yu Song
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.
| | - Xincen Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Yanjun Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.
| | - Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China.
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China; National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs and Biological Products, Qingdao, Shandong, China.
| |
Collapse
|
7
|
Gunasekar SK, Heebink J, Carpenter DH, Kumar A, Xie L, Zhang H, Schilling JD, Sah R. Adipose-targeted SWELL1 deletion exacerbates obesity- and age-related nonalcoholic fatty liver disease. JCI Insight 2023; 8:e154940. [PMID: 36749637 PMCID: PMC10077479 DOI: 10.1172/jci.insight.154940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Healthy expansion of adipose tissue is critical for the maintenance of metabolic health, providing an optimized reservoir for energy storage in the form of triacylglycerol-rich lipoproteins. Dysfunctional adipocytes that are unable to efficiently store lipid can result in lipodystrophy and contribute to nonalcoholic fatty liver disease (NAFLD) and metabolic syndrome. Leucine-rich repeat containing protein 8a/SWELL1 functionally encodes the volume-regulated anion channel complex in adipocytes, is induced in early obesity, and is required for normal adipocyte expansion during high-fat feeding. Adipose-specific SWELL1 ablation (Adipo KO) leads to insulin resistance and hyperglycemia during caloric excess, both of which are associated with NAFLD. Here, we show that Adipo-KO mice exhibited impaired adipose depot expansion and excess lipolysis when raised on a variety of high-fat diets, resulting in increased diacylglycerides and hepatic steatosis, thereby driving liver injury. Liver lipidomic analysis revealed increases in oleic acid-containing hepatic triacylglycerides and injurious hepatic diacylglyceride species, with reductions in hepatocyte-protective phospholipids and antiinflammatory free fatty acids. Aged Adipo-KO mice developed hepatic steatosis on a regular chow diet, and Adipo-KO male mice developed spontaneous, aggressive hepatocellular carcinomas (HCCs). These data highlight the importance of adipocyte SWELL1 for healthy adipocyte expansion to protect against NAFLD and HCC in the setting of overnutrition and with aging.
Collapse
Affiliation(s)
- Susheel K. Gunasekar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Heebink
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Danielle H. Carpenter
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Litao Xie
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haixia Zhang
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joel D. Schilling
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Hu G, Ling C, Chi L, Thind MK, Furse S, Koulman A, Swann JR, Lee D, Calon MM, Bourdon C, Versloot CJ, Bakker BM, Gonzales GB, Kim PK, Bandsma RHJ. The role of the tryptophan-NAD + pathway in a mouse model of severe malnutrition induced liver dysfunction. Nat Commun 2022; 13:7576. [PMID: 36481684 PMCID: PMC9732354 DOI: 10.1038/s41467-022-35317-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Mortality in children with severe malnutrition is strongly related to signs of metabolic dysfunction, such as hypoglycemia. Lower circulating tryptophan levels in children with severe malnutrition suggest a possible disturbance in the tryptophan-nicotinamide adenine dinucleotide (TRP-NAD+) pathway and subsequently in NAD+ dependent metabolism regulator sirtuin1 (SIRT1). Here we show that severe malnutrition in weanling mice, induced by 2-weeks of low protein diet feeding from weaning, leads to an impaired TRP-NAD+ pathway with decreased NAD+ levels and affects hepatic mitochondrial turnover and function. We demonstrate that stimulating the TRP-NAD+ pathway with NAD+ precursors improves hepatic mitochondrial and overall metabolic function through SIRT1 modulation. Activating SIRT1 is sufficient to induce improvement in metabolic functions. Our findings indicate that modulating the TRP-NAD+ pathway can improve liver metabolic function in a mouse model of severe malnutrition. These results could lead to the development of new interventions for children with severe malnutrition.
Collapse
Affiliation(s)
- Guanlan Hu
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Catriona Ling
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Lijun Chi
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Mehakpreet K. Thind
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Samuel Furse
- grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-Metabolic Research Laboratories, Institute of Metabolic Sciences, University of Cambridge, CB2 0QQ Cambridge, UK ,grid.4903.e0000 0001 2097 4353Biological Chemistry Group, Royal Botanic Gardens, Kew, Kew Green, TW9 3AE Richmond, UK
| | - Albert Koulman
- grid.5335.00000000121885934Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-Metabolic Research Laboratories, Institute of Metabolic Sciences, University of Cambridge, CB2 0QQ Cambridge, UK
| | - Jonathan R. Swann
- grid.5491.90000 0004 1936 9297School of Human Development and Health, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK ,grid.7445.20000 0001 2113 8111Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, SW7 2AZ London, UK
| | - Dorothy Lee
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Marjolein M. Calon
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Celine Bourdon
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.511677.3The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Christian J. Versloot
- grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara M. Bakker
- grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Bryan Gonzales
- grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.4818.50000 0001 0791 5666Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Peter K. Kim
- grid.17063.330000 0001 2157 2938Department of Biochemistry, University of Toronto, M5S 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Cell Biology Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| | - Robert H. J. Bandsma
- grid.17063.330000 0001 2157 2938Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, M5G 1A8 Toronto, Canada ,grid.42327.300000 0004 0473 9646Translational Medicine Program, The Hospital for Sick Children, M5G 0A4 Toronto, Canada ,grid.511677.3The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya ,grid.4494.d0000 0000 9558 4598Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.42327.300000 0004 0473 9646Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, M5G 0A4 Toronto, Canada
| |
Collapse
|
9
|
Ai ZL, Zhang X, Ge W, Zhong YB, Wang HY, Zuo ZY, Liu DY. Salvia miltiorrhiza extract may exert an anti-obesity effect in rats with high-fat diet-induced obesity by modulating gut microbiome and lipid metabolism. World J Gastroenterol 2022; 28:6131-6156. [PMID: 36483153 PMCID: PMC9724488 DOI: 10.3748/wjg.v28.i43.6131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/21/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Studies have shown that a high-fat diet (HFD) can alter gut microbiota (GM) homeostasis and participate in lipid metabolism disorders associated with obesity. Therefore, regulating the construction of GM with the balance of lipid metabolism has become essential for treating obesity. Salvia miltiorrhiza extract (Sal), a common traditional Chinese medicine, has been proven effective against atherosclerosis, hyperlipidemia, obesity, and other dyslipidemia-related diseases. AIM To investigate the anti-obesity effects of Sal in rats with HFD-induced obesity, and explore the underlying mechanism by focusing on GM and lipid metabolism. METHODS Obesity was induced in rats with an HFD for 7 wk, and Sal (0.675 g/1.35 g/2.70 g/kg/d) was administered to treat obese rats for 8 wk. The therapeutic effect was evaluated by body weight, body fat index, waistline, and serum lipid level. Lipid factors (cAMP, PKA, and HSL) in liver and fat homogenates were analyzed by ELISA. The effect of Sal on GM and lipid metabolism was assessed by 16S rRNA-based microbiota analysis and untargeted lipidomic analysis (LC-MS/MS), respectively. RESULTS Sal treatment markedly reduced weight, body fat index, serum triglycerides (TG), total cholesterol (TC), low-density lipoprotein, glucose, free fatty acid, hepatic lipid accumulation, and adipocyte vacuolation, and increased serum high-density lipoprotein (HDL-C) in rats with HFD-induced obesity. These effects were associated with increased concentrations of lipid factors such as cAMP, PKA, and HSL in the liver and adipose tissues, enhanced gut integrity, and improved lipid metabolism. GM analysis revealed that Sal could reverse HFD-induced dysbacteriosis by promoting the abundance of Actinobacteriota and Proteobacteria, and decreasing the growth of Firmicutes and Desulfobacterita. Furthermore, LC-MS/MS analysis indicated that Sal decreased TGs (TG18:2/18:2/20:4, TG16:0/18:2/22:6), DGs (DG14:0/22:6, DG22:6/22:6), CL (18:2/ 18:1/18:1/20:0), and increased ceramides (Cers; Cer d16:0/21:0, Cer d16:1/24:1), (O-acyl)-ω-hydroxy fatty acids (OAHFAs; OAHFA18:0/14:0) in the feces of rats. Spearman's correlation analysis further indicated that TGs, DGs, and CL were negatively related to the abundance of Facklamia and Dubosiella, and positively correlated with Blautia and Quinella, while OAHFAs and Cers were the opposite. CONCLUSION Sal has an anti-obesity effect by regulating the GM and lipid metabolism.
Collapse
Affiliation(s)
- Zi-Li Ai
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xian Zhang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wei Ge
- Department of Proctology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - You-Bao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Yan Wang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Zheng-Yun Zuo
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| |
Collapse
|
10
|
Pan Z, Mao B, Zhang Q, Tang X, Yang B, Zhao J, Cui S, Zhang H. Postbiotics Prepared Using Lactobacillus paracasei CCFM1224 Prevent Nonalcoholic Fatty Liver Disease by Modulating the Gut Microbiota and Liver Metabolism. Int J Mol Sci 2022; 23:ijms232113522. [PMID: 36362307 PMCID: PMC9653709 DOI: 10.3390/ijms232113522] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Postbiotics are rich in a variety of bioactive components, which may have beneficial effects in inhibiting hepatic lipid accumulation. In this study, we investigated the preventive effects of postbiotics (POST) prepared from Lactobacillus paracasei on non-alcoholic fatty liver disease (NAFLD). Our results showed that when mice ingested a high-fat diet (HFD) and POST simultaneously, weight gain was slowed, epididymal white fat hypertrophy and insulin resistance were suppressed, serum biochemical indicators related to blood lipid metabolism were improved, and hepatic steatosis and liver inflammation decreased. Bacterial sequencing showed that POST modulated the gut microbiota in HFD mice, increasing the relative abundance of Akkermansia and reducing the relative abundance of Lachnospiraceae NK4A136 group, Ruminiclostridium and Bilophila. Spearman’s correlation analysis revealed significant correlations between lipid metabolism parameters and gut microbes. Functional prediction results showed that the regulation of gut microbiota was associated with the improvement of metabolic status. The metabolomic analysis of the liver revealed that POST-regulated liver metabolic pathways, such as glycerophospholipid and ether lipid metabolism, pantothenate and CoA biosynthesis, some parts of amino acid metabolism, and other metabolic pathways. In addition, POST regulated the gene expression in hepatocytes at the mRNA level, thereby regulating lipid metabolism. These findings suggest that POST plays a protective role against NAFLD and may exert its efficacy by modulating the gut microbiota and liver metabolism, and these findings may be applied to related functional foods.
Collapse
Affiliation(s)
- Zhenghao Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Correspondence: ; Tel.: +86-0510-85912155
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
11
|
Nagumalli SK, Willett RA, de Conti A, Tryndyak VP, Avigan MI, da Costa GG, Beland FA, Rusyn I, Pogribny IP. Lipidomic profiling of the hepatic esterified fatty acid composition in diet-induced nonalcoholic fatty liver disease in genetically diverse Collaborative Cross mice. J Nutr Biochem 2022; 109:109108. [PMID: 35858665 PMCID: PMC10103579 DOI: 10.1016/j.jnutbio.2022.109108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 04/06/2022] [Accepted: 06/20/2022] [Indexed: 01/24/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), one of the most common forms of chronic liver disease, is characterized by the excessive accumulation of lipid species in hepatocytes. Recent studies have indicated that in addition to the total lipid quantities, changes in lipid composition are a determining factor in hepatic lipotoxicity. Using ultra-high performance liquid chromatography coupled with electrospray tandem mass spectrometry, we analyzed the esterified fatty acid composition in 24 strains of male and female Collaborative Cross (CC) mice fed a high fat/high sucrose (HF/HS) diet for 12 weeks. Changes in lipid composition were found in all strains after the HF/HS diet, most notably characterized by increases in monounsaturated fatty acids (MUFA) and decreases in polyunsaturated fatty acids (PUFA). Similar changes in MUFA and PUFA were observed in a choline- and folate-deficient (CFD) mouse model of NAFLD, as well as in hepatocytes treated in vitro with free fatty acids. Analysis of fatty acid composition revealed that alterations were accompanied by an increase in the estimated activity of MUFA generating SCD1 enzyme and an estimated decrease in the activity of PUFA generating FADS1 and FADS2 enzymes. PUFA/MUFA ratios were inversely correlated with lipid accumulation in male and female CC mice fed the HF/HS diet and with morphological markers of hepatic injury in CFD diet-fed mouse model of NAFLD. These results demonstrate that different models of NAFLD are characterized by similar changes in the esterified fatty acid composition and that alterations in PUFA/MUFA ratios may serve as a diagnostic marker for NAFLD severity.
Collapse
Affiliation(s)
- Suresh K Nagumalli
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Rose A Willett
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Aline de Conti
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Volodymyr P Tryndyak
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Mark I Avigan
- Office of Pharmacovigilance and Epidemiology, FDA-Center for Drug Evaluation and Research, Silver Spring, Maryland, USA
| | - Gonçalo Gamboa da Costa
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Igor P Pogribny
- Division of Biochemical Toxicology, FDA-National Center for Toxicological Research, Jefferson, Arkansas, USA.
| |
Collapse
|
12
|
Huang M, Koizumi A, Narita S, Nakanishi H, Sato H, Kashima S, Nara T, Kanda S, Numakura K, Saito M, Satoh S, Nanjo H, Sasaki T, Habuchi T. Altering phosphoinositides in high-fat diet-associated prostate tumor xenograft growth. MedComm (Beijing) 2021; 2:756-764. [PMID: 34977875 PMCID: PMC8706770 DOI: 10.1002/mco2.89] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/08/2022] Open
Abstract
The metabolic reprogramming of phospholipids may affect intracellular signal transduction pathways. A high-fat diet (HFD) is attributed to prostate cancer (PCa) progression, but the expression pattern and role of phospholipids in HFD-mediated PCa progression remains unclear. In this study, HFD enhanced LNCaP xenograft tumor growth by upregulating the phosphatidylinositol (PI) 3-kinase (PI3K)/AKT signaling pathway. A lipidomic analysis using xenograft tumors showed that phosphoinositides, especially PI (3,4,5)-trisphosphate (PIP3), including several species containing C38:4, C38:3, and C40:4 fatty acids, increased in the HFD group compared to control. Fatty acid synthase (FASN) was significantly upregulated in xenograft tumors under HFD in both gene and protein levels. PCa cell growth was significantly inhibited through the decreased AKT signaling pathway by treatment with cerulenin, a chemical FASN inhibitor, which also downregulated PIP, PIP2, and PIP3 but not PI. Thus, dietary fat influences PCa progression and alters phosphoinositides, especially PIP3, a critical player in the PI3K/AKT pathway. These results may offer appropriate targets, such as FASN, for dietary intervention and/or chemoprevention to reduce PCa incidence and progression.
Collapse
Affiliation(s)
- Mingguo Huang
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Atsushi Koizumi
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Shintaro Narita
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Hiroki Nakanishi
- Research Center for BiosignalAkita University Graduate School of MedicineAkitaJapan
| | - Hiromi Sato
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Soki Kashima
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Taketoshi Nara
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Sohei Kanda
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Kazuyuki Numakura
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Mitsuru Saito
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Shigeru Satoh
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| | - Hiroshi Nanjo
- Department of Clinical PathologyAkita University Graduate School of MedicineAkitaJapan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental UniversityBunkyo‐kuTokyoJapan
| | - Tomonori Habuchi
- Department of UrologyAkita University Graduate School of MedicineAkitaJapan
| |
Collapse
|
13
|
Koelmel JP, Aristizabal-Henao JJ, Ni Z, Fedorova M, Kato S, Otoki Y, Nakagawa K, Lin EZ, Godri Pollitt KJ, Vasiliou V, Guingab JD, Garrett TJ, Williams TL, Bowden JA, Penumetcha M. A Novel Technique for Redox Lipidomics Using Mass Spectrometry: Application on Vegetable Oils Used to Fry Potatoes. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1798-1809. [PMID: 34096708 DOI: 10.1021/jasms.1c00150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Vegetables oils, rich in polyunsaturated fatty acids, are vulnerable to oxidation during manufacturing, processing, and food preparation. Currently, individual oxidation products are not well characterized, and hence, the health impacts of these unique lipid species remain unknown. Here, we introduce an extensive oxidized lipidomics in silico tandem mass spectrometry library and integrate these libraries within a user-friendly software covering a comprehensive redox lipidomics workflow. We apply this workflow to olive, soy, and walnut cooking oil; comparing unheated oil, oil after deep frying potatoes, and oil after oven frying potatoes. We annotated over a thousand oxidized triglycerides across 273 features (many coeluted). This software was validated against traditional chemical assays of oxidation, known oxidized lipids in castor oil, synthesized standards, and an alternate software LPPtiger. Development of these new software programs for redox lipidomics opens the door to characterize health implications of individual oxidation products.
Collapse
Affiliation(s)
- Jeremy P Koelmel
- School of Public Health, Yale University, New Haven, Connecticut, 06520, United States
| | - Juan J Aristizabal-Henao
- Center for Environmental and Human Toxicology & Department of Physiological Sciences, University of Florida, Gainesville, Florida 32608, United States
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig 01403Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, 04103, Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig 01403Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, 04103, Germany
| | - Shunji Kato
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan
| | - Yurika Otoki
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan
| | - Elizabeth Z Lin
- School of Public Health, Yale University, New Haven, Connecticut, 06520, United States
| | | | - Vasilis Vasiliou
- School of Public Health, Yale University, New Haven, Connecticut, 06520, United States
| | - Joy D Guingab
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, United States
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Traycie L Williams
- School of Nutrition, Kinesiology and Psychological Science, University of Central Missouri, Warrensburg, Missouri 64093, United States
| | - John A Bowden
- Center for Environmental and Human Toxicology & Department of Physiological Sciences, University of Florida, Gainesville, Florida 32608, United States
- Department of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Meera Penumetcha
- School of Nutrition, Kinesiology and Psychological Science, University of Central Missouri, Warrensburg, Missouri 64093, United States
| |
Collapse
|
14
|
Bramlage KS, Bhattacharjee J, Kirby M, Myronovych A, Gupta R, Gonzalez RMS, Xanthakos S, Bove K, Kohli R. A Diet High in Fat and Fructose Induces Early Hepatic Mitochondrial Aging. J Pediatr Gastroenterol Nutr 2021; 73:99-102. [PMID: 34135298 PMCID: PMC8549102 DOI: 10.1097/mpg.0000000000003068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT To investigate the effect of high fructose diet on ultrastructure and content of hepatic mitochondria, we randomized 6-8 weeks old male C57Bl6/J mice to ad lib chow or high-fat-high-fructose (HF2) diet for 32 weeks. HF2-fed mice gained more weight, had higher plasma alanine aminotransferase, and fasting glucose levels and increased hepatic triglyceride content at all time points compared to chow-fed mice. HF2-fed mice had lower mitochondrial to nuclear DNA ratio compared to chow-fed mice. HF2-fed mice had lower average mitochondrial surface area and the number of mitochondria compared to chow-fed mice. HF2-fed mice had higher expression of the hepatic endoplasmic reticulum stress marker Chop, compared to chow-fed mice. A diet high in fat and fructose leads to enhanced hepatic mitochondrial aging, depletion, and dysfunction, which may be important determinants of nonalcoholic steatohepatitis pathogenesis.
Collapse
Affiliation(s)
- Kristin S. Bramlage
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jashdeep Bhattacharjee
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Michelle Kirby
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Andriy Myronovych
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Rohun Gupta
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| | | | - Stavra Xanthakos
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kevin Bove
- Department of Pediatrics, Division of Pathology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| |
Collapse
|
15
|
Changes in Key Mitochondrial Lipids Accompany Mitochondrial Dysfunction and Oxidative Stress in NAFLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9986299. [PMID: 34257827 PMCID: PMC8257344 DOI: 10.1155/2021/9986299] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/06/2021] [Indexed: 12/30/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a dysmetabolic hepatic damage of increasing severity: simple fat accumulation (steatosis), nonalcoholic steatohepatitis (NASH), and hepatic fibrosis. Oxidative stress is considered an important factor in producing hepatocyte injury associated with NAFLD progression. Studies also suggest a link between the accumulation of specific hepatic lipid species, mitochondrial dysfunction, and the progression of NAFLD. However, it is unclear whether mitochondrial lipid modifications are involved in NAFLD progression. To gain insight into the relationship between mitochondrial lipids and disease progression through different stages of NAFLD, we performed lipidomic analyses on mouse livers at different stages of western diet-induced NAFLD, with or without hepatic fibrosis. After organelle separation, we studied separately the mitochondrial and the “nonmitochondrial” hepatic lipidomes. We identified 719 lipid species from 16 lipid families. Remarkably, the western diet triggered time-dependent changes in the mitochondrial lipidome, whereas the “nonmitochondrial” lipidome showed little difference with levels of hepatic steatosis or the presence of fibrosis. In mitochondria, the changes in the lipidome preceded hepatic fibrosis. In particular, two critical phospholipids, phosphatidic acid (PA) and cardiolipin (CL), displayed opposite responses in mitochondria. Decrease in CL and increase in PA were concurrent with an increase of coenzyme Q. Electron paramagnetic resonance spectroscopy superoxide spin trapping and Cu2+ measurement showed the progressive increase in oxidative stress in the liver. Overall, these results suggest mitochondrial lipid modifications could act as an early event in mitochondrial dysfunction and NAFLD progression.
Collapse
|
16
|
Zhou J, Zhang Y, Wu J, Qiao M, Xu Z, Peng X, Mei S. Proteomic and lipidomic analyses reveal saturated fatty acids, phosphatidylinositol, phosphatidylserine, and associated proteins contributing to intramuscular fat deposition. J Proteomics 2021; 241:104235. [PMID: 33894376 DOI: 10.1016/j.jprot.2021.104235] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
Intramuscular fat (IMF) content is an important factor in porcine meat quality. Previous studies have screened multiple candidate genes related to IMF deposition, but the lipids that affect IMF deposition and their lipid-protein network remain unknown. In this study, we performed proteomic and lipidomic analyses of the longissimus dorsi (LD) muscle from high-IMF (IMFH) and low-IMF (IMF-L) groups of Xidu black pigs. Eighty-eight proteins and 143 lipids were differentially abundant between the groups. The differentially abundant proteins were found to be involved in cholesterol metabolism, the PPAR signaling pathway, and ferroptosis. The triacylglycerols (TAGs) upregulated in the IMF-H group were mainly shown to be synthesized by saturated fatty acids (SFAs), while the downregulated TAGs were mainly synthesized by polyunsaturated fatty acids (PUFAs). All differentially abundant phosphatidylinositols (PIs) and phosphatidylserines (PSs) were found to be upregulated in the IMF-H group. A correlation analysis of the proteomic and lipidomic revealed candidate proteins (APOA4, VDAC3, PRNP, CTSB, GSPT1) related to TAG, PI, and PS lipids. These results revealed differences in proteins and lipids between the IMF-H and IMF-L groups, which represent new candidate proteins and lipids that should be investigated to determine the molecular mechanisms controlling IMF deposition in pigs. SIGNIFICANCE: Intramuscular fat (IMF) is a key factor affecting meat quality, and meat with a higher IMF content can have a better flavor. In this study, proteomic results show that the ferroptosis pathway, including the PRNP, VDAC3 and CP proteins, affects IMF deposition. Lipid composition is the key factor affecting IMF deposition, but there are few reports on this. In this study, through lipidomic analysis, we suggest that saturated fatty acid (SFA), phosphatidylinositol (PI), and phosphatidylserine (PS) may contribute to IMF deposition. A correlation analysis reveals the potential regulatory network between lipids and proteins. This study clarifies the difference in protein and lipid compositions in longissimus dorsi (LD) muscle with high and low IMF contents. This information suggests that it would be beneficial to increase the intramuscular fat content of pork not only from a genetic perspective but also from a nutritional perspective.
Collapse
Affiliation(s)
- Jiawei Zhou
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Yu Zhang
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Junjing Wu
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Mu Qiao
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Zhong Xu
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Xianwen Peng
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Shuqi Mei
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China.
| |
Collapse
|
17
|
Tran A, Wan L, Xu Z, Haro JM, Li B, Jones JW. Lithium Hydroxide Hydrolysis Combined with MALDI TOF Mass Spectrometry for Rapid Sphingolipid Detection. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:289-300. [PMID: 33124427 PMCID: PMC7790884 DOI: 10.1021/jasms.0c00322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Sphingolipids have diverse structural and bioactive functions that play important roles in many key biological processes. Factors such as low relative abundance, varied structures, and a dynamic concentration range provide a difficult analytical challenge for sphingolipid detection. To further improve mass-spectrometry-based sphingolipid analysis, lithium adduct consolidation was implemented to decrease spectral complexity and combine signal intensities, leading to increased specificity and sensitivity. We report the use of lithium hydroxide as a base in a routine hydrolysis procedure in order to effectively remove common ionization suppressants (such as glycolipids and glycerophospholipids) and introduce a source of lithium into the sample. In conjunction, an optimized MALDI matrix system, featuring 2',4',6'-trihydroxyacetophenone (THAP) is used to facilitate lithium adduct consolidation during the MALDI process. The result is a robust and high-throughput sphingolipid detection scheme, particularly of low-abundance ceramides. Application of our developed workflow includes the detection of differentially expressed liver sphingolipid profiles from a high-fat-induced obesity mouse model. We also demonstrate the method's effectiveness in detecting various sphingolipids in brain and plasma matrices. These results were corroborated with data from UHPLC HR MS/MS and MALDI FT-ICR, verifying the efficacy of the method application. Overall, we demonstrate a high-throughput workflow for sphingolipid analysis in various biological matrices by the use of MALDI TOF and lithium adduct consolidation.
Collapse
Affiliation(s)
- Anh Tran
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Liting Wan
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland 21201, United States
| | - Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou 510640, China
| | - Janette M Haro
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland 21201, United States
| | - Bing Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
18
|
Streidl T, Karkossa I, Segura Muñoz RR, Eberl C, Zaufel A, Plagge J, Schmaltz R, Schubert K, Basic M, Schneider KM, Afify M, Trautwein C, Tolba R, Stecher B, Doden HL, Ridlon JM, Ecker J, Moustafa T, von Bergen M, Ramer-Tait AE, Clavel T. The gut bacterium Extibacter muris produces secondary bile acids and influences liver physiology in gnotobiotic mice. Gut Microbes 2021; 13:1-21. [PMID: 33382950 PMCID: PMC7781625 DOI: 10.1080/19490976.2020.1854008] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/09/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
Extibacter muris is a newly described mouse gut bacterium which metabolizes cholic acid (CA) to deoxycholic acid (DCA) via 7α-dehydroxylation. Although bile acids influence metabolic and inflammatory responses, few in vivo models exist for studying their metabolism and impact on the host. Mice were colonized from birth with the simplified community Oligo-MM12 with or without E. muris. As the metabolism of bile acids is known to affect lipid homeostasis, mice were fed either a low- or high-fat diet for eight weeks before sampling and analyses targeting the gut and liver. Multiple Oligo-MM12 strains were capable of deconjugating primary bile acids in vitro. E. muris produced DCA from CA either as pure compound or in mouse bile. This production was inducible by CA in vitro. Ursodeoxycholic, chenodeoxycholic, and β-muricholic acid were not metabolized under the conditions tested. All gnotobiotic mice were stably colonized with E. muris, which showed higher relative abundances after HF diet feeding. The presence of E. muris had minor, diet-dependent effects on Oligo-MM12 communities. The secondary bile acids DCA and surprisingly LCA and their taurine conjugates were detected exclusively in E. muris-colonized mice. E. muris colonization did not influence body weight, white adipose tissue mass, liver histopathology, hepatic aspartate aminotransferase, or blood levels of cholesterol, insulin, and paralytic peptide (PP). However, proteomics revealed shifts in hepatic pathways involved in amino acid, glucose, lipid, energy, and drug metabolism in E. muris-colonized mice. Liver fatty acid composition was substantially altered by dietary fat but not by E. muris.In summary, E. muris stably colonized the gut of mice harboring a simplified community and produced secondary bile acids, which affected proteomes in the liver. This new gnotobiotic mouse model can now be used to study the pathophysiological role of secondary bile acids in vivo.
Collapse
Affiliation(s)
- Theresa Streidl
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH, Aachen, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research, Leipzig, Germany
| | | | - Claudia Eberl
- Max Von Pettenkofer Institute of Hygiene and Medical Microbiology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alex Zaufel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University, Graz, Austria
| | - Johannes Plagge
- Research Group Lipid Metabolism, ZIEL Institute for Food & Health, Technical University, Munich, Germany
| | - Robert Schmaltz
- Department of Food Science & Technology, University of Nebraska-Lincoln, NE, USA
| | - Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research, Leipzig, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Kai Markus Schneider
- Department of Internal Medicine III, University Hospital of RWTH, Aachen, Germany
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mamdouh Afify
- Institute for Laboratory Animal Science, Faculty of Medicine, University Hospital of RWTH, Aachen, Germany
- Clinic for Cardiology (Internal Medicine I), University Hospital of RWTH, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital of RWTH, Aachen, Germany
| | - René Tolba
- Institute for Laboratory Animal Science, Faculty of Medicine, University Hospital of RWTH, Aachen, Germany
| | - Bärbel Stecher
- Max Von Pettenkofer Institute of Hygiene and Medical Microbiology, Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Infection Research (DZIF); Partner Site Munich, Munich, Germany
| | - Heidi L. Doden
- Microbiome Metabolic Engineering Theme, Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jason M. Ridlon
- Microbiome Metabolic Engineering Theme, Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Josef Ecker
- Research Group Lipid Metabolism, ZIEL Institute for Food & Health, Technical University, Munich, Germany
| | - Tarek Moustafa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University, Graz, Austria
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research, Leipzig, Germany
- Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Amanda E. Ramer-Tait
- Department of Food Science & Technology, University of Nebraska-Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Hannover, NE, USA
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH, Aachen, Germany
| |
Collapse
|
19
|
Haberl EM, Pohl R, Rein-Fischboeck L, Höring M, Krautbauer S, Liebisch G, Buechler C. Hepatic lipid profile in mice fed a choline-deficient, low-methionine diet resembles human non-alcoholic fatty liver disease. Lipids Health Dis 2020; 19:250. [PMID: 33298075 PMCID: PMC7727224 DOI: 10.1186/s12944-020-01425-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Emerging data support a role for lipids in non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) in humans. With experimental models such data can be challenged or validated. Mice fed a low-methionine, choline-deficient (LMCD) diet develop NASH and, when injected with diethylnitrosamine (DEN), HCC. Here, lipidomic analysis was used to elucidate whether the NASH and HCC associated lipid derangements resemble the lipid profile of the human disease. METHODS Lipids were measured in the liver of mice fed a control or a LMCD diet for 16 weeks. DEN was injected at young age to initiate hepatocarcinogenesis. DEN treatment associated changes of the lipid composition and the tumor lipidome were evaluated. RESULTS LMCD diet fed mice accumulated ceramides and triacylglycerols in the liver. Phospholipids enriched with monounsaturated fatty acids were also increased, whereas hepatic cholesterol levels remained unchanged in the LMCD model. Phosphatidylcholine and lysophosphatidylcholine concentrations declined in the liver of LMCD diet fed mice. The changes of most lipids associated with LMCD diet feeding were similar between water and DEN injected mice. Several polyunsaturated (PU) diacylglycerol species were already low in the liver of DEN injected mice fed the control diet. Tumors developed in the liver of LMCD diet fed mice injected with DEN. The tumor specific lipid profile, however, did not resemble the decrease of ceramides and PU phospholipids, which was consistently described in human HCC. Triacylglycerols declined in the cancer tissues, which is in accordance with a low expression of lipogenic enzymes in the tumors. CONCLUSIONS The LMCD model is suitable to study NASH associated lipid reprogramming. Hepatic lipid profile was modestly modified in the DEN injected mice suggesting a function of these derangements in carcinogenesis. Lipid composition of liver tumors did not resemble the human HCC lipidome, and most notably, lipogenesis and triacylglycerol levels were suppressed.
Collapse
Affiliation(s)
- Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany.
| |
Collapse
|
20
|
Li L, Wang H, Jones JW. Sphingolipid metabolism as a marker of hepatotoxicity in drug-induced liver injury. Prostaglandins Other Lipid Mediat 2020; 151:106484. [PMID: 33007444 PMCID: PMC7669681 DOI: 10.1016/j.prostaglandins.2020.106484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/09/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) has a substantial impact on human health and is a major monetary burden on the drug development process. Presently, there is a lack of robust and analytically validated markers for predicting and early diagnosis of DILI. Sphingolipid metabolism and subsequent disruption of sphingolipid homeostasis has been documented to play a key role contributing to hepatocellular death and subsequent liver injury. A more comprehensive understanding of sphingolipid metabolism in response to liver toxicity has great potential to gain mechanistic insight into hepatotoxicity and define molecular markers that are responsible for hepatocyte dysfunction. Here, we present an analytical platform that provides multidimensional mass spectrometry-based datasets for comprehensive structure characterization of sphingolipids extracted from human primary hepatocytes (HPH) exposed to toxic levels of acetaminophen (APAP). Sphingolipid metabolism as measured by characterization of individual sphingolipid structure was sensitive to APAP toxicity displaying a concentration-dependent response. A number of sphingolipid structures were differentially expressed across varying APAP exposures highlighting the unique role sphingolipid metabolism has in response to hepatotoxicity and its potential use as a molecular marker in DILI.
Collapse
Affiliation(s)
- Linhao Li
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Hongbing Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Jace W Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States.
| |
Collapse
|
21
|
Liu Z, Zhu H, Wang W, Xu J, Que S, Zhuang L, Qian J, Wang S, Yu J, Zhang F, Yin S, Xie H, Zhou L, Geng L, Zheng S. Metabonomic Profile of Macrosteatotic Allografts for Orthotopic Liver Transplantation in Patients With Initial Poor Function: Mechanistic Investigation and Prognostic Prediction. Front Cell Dev Biol 2020; 8:826. [PMID: 32984324 PMCID: PMC7484052 DOI: 10.3389/fcell.2020.00826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Our previous study revealled amplified hazardous effects of macrosteatosis (MaS) on graft failure (GF) in recipients with severe liver damage in short post-operative days, with vague mechanism inside. AIM We aimed to uncover the molecular mechanism of donor MaS on GF, and construct the predictive model to monitor post-transplant prognosis based on "omics" perspective. METHODS Ultra-performance liquid chromatography coupled to mass spectrometry metabolomic analysis was performed in allograft tissues from 82 patients with initial poor function (IPF) from multi-liver transplant (LT) centers. Pathway analysis was performed by on-line toolkit Metaboanalyst (v 3.0). Predictive model was constructed based on combinative metabonomic and clinical data extracted by stepwised cox proportional analysis. RESULTS Principle component analysis (PCA) analysis revealled stratification on metabolic feature in organs classified by MaS status. Differential metabolits both associated with MaS and GF were significantly enriched on pathway of glycerophospholipid metabolism (P < 0.05). Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) involved in glycerophospholipid metabolism was significantly decreased in cases with MaS donors and GF (P < 0.05). Better prediction was observed on graft survival by combinative model (area under the curve = 0.91) and confirmed by internal validation. CONCLUSION Metabonomic features of allografts can be clearly distinguished by MaS status in patients with IPF. Dysfunction on glycerophospholipid metabolism was culprit to link donor MaS and final GF. Decrement on PC and PE exerted the fatal effects of MaS on organ failure. Metabonomic data might help for monitoring long-term graft survival after LT.
Collapse
Affiliation(s)
- Zhengtao Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hai Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenchao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Li Zhuang
- Shulan Hospital (Hangzhou), Hangzhou, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Zhang
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengyong Yin
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyang Xie
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lei Geng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Shulan Hospital (Hangzhou), Hangzhou, China
| |
Collapse
|
22
|
Hasanpour M, Iranshahy M, Iranshahi M. The application of metabolomics in investigating anti-diabetic activity of medicinal plants. Biomed Pharmacother 2020; 128:110263. [DOI: 10.1016/j.biopha.2020.110263] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022] Open
|
23
|
GOWDA SGB, GAO ZJ, CHEN Z, ABE T, HORI S, FUKIYA S, ISHIZUKA S, YOKOTA A, CHIBA H, HUI SP. Untargeted Lipidomic Analysis of Plasma from High-fat Diet-induced Obese Rats Using UHPLC–Linear Trap Quadrupole–Orbitrap MS. ANAL SCI 2020; 36:821-828. [DOI: 10.2116/analsci.19p442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
| | - Zi-Jun GAO
- Faculty of Health Sciences, Hokkaido University
| | - Zhen CHEN
- Faculty of Health Sciences, Hokkaido University
| | - Takayuki ABE
- Graduate School of Agriculture, Hokkaido University
| | - Shota HORI
- Graduate School of Agriculture, Hokkaido University
| | | | | | | | - Hitoshi CHIBA
- Department of Nutrition, Sapporo University of Health Sciences
| | | |
Collapse
|
24
|
O’Dwyer C, Yaworski R, Katsumura S, Ghorbani P, Gobeil Odai K, Nunes JR, LeBlond ND, Sanjana S, Smith TT, Han S, Margison KD, Alain T, Morita M, Fullerton MD. Hepatic Choline Transport Is Inhibited During Fatty Acid-Induced Lipotoxicity and Obesity. Hepatol Commun 2020; 4:876-889. [PMID: 32490323 PMCID: PMC7262319 DOI: 10.1002/hep4.1516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 01/11/2023] Open
Abstract
Choline is an essential nutrient and a critical component of the membrane phospholipid phosphatidylcholine (PC), the neurotransmitter acetylcholine, while also contributing to the methylation pathway. In the liver specifically, PC is the major membrane constituent and can be synthesized by the cytidine diphosphate-choline or the phosphatidylethanolamine N-methyltransferase pathway. With the continuing global rise in the rates of obesity and nonalcoholic fatty liver disease, we sought to explore how excess fatty acids on primary hepatocytes and diet-induced obesity affect choline uptake and metabolism. Our results demonstrate that hepatocytes chronically treated with palmitate, but not oleate or a mixture, had decreased choline uptake, which was associated with lower choline incorporation into PC and lower expression of choline transport proteins. Interestingly, a reduction in the rate of degradation spared PC levels in response to palmitate when compared with control. The effects of palmitate treatment were independent of endoplasmic reticulum stress, which counterintuitively augmented choline transport and transporter expression. In a model of obesity-induced hepatic steatosis, male mice fed a 60% high-fat diet for 10 weeks had significantly diminished hepatic choline uptake compared with lean mice fed a control diet. Although the transcript and protein expression of various choline metabolic enzymes fluctuated slightly, we observed reduced protein expression of choline transporter-like 1 (CTL1) in the liver of mice fed a high-fat diet. Polysome profile analyses revealed that in livers of obese mice, the CTL1 transcript, despite being more abundant, was translated to a lesser extent compared with lean controls. Finally, human liver cells demonstrated a similar response to palmitate treatment. Conclusion: Our results suggest that the altered fatty acid milieu seen in obesity-induced fatty liver disease progression may adversely affect choline metabolism, potentially through CTL1, but that compensatory mechanisms work to maintain phospholipid homeostasis.
Collapse
Affiliation(s)
- Conor O’Dwyer
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Rebecca Yaworski
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Sakie Katsumura
- Department of Molecular MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTX
| | - Peyman Ghorbani
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Kaelan Gobeil Odai
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Julia R.C. Nunes
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Nicholas D. LeBlond
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Sabrin Sanjana
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Tyler T.K. Smith
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Shauna Han
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Kaitlyn D. Margison
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
- Children’s Hospital of Eastern Ontario Research InstituteOttawaONCanada
| | - Masahiro Morita
- Department of Molecular MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTX
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTX
- Institute of Resource Development and AnalysisKumamoto UniversityKumamotoJapan
| | - Morgan D. Fullerton
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for InfectionImmunity and Inflammation and Centre for Catalysis Research and InnovationUniversity of OttawaOttawaONCanada
| |
Collapse
|
25
|
Sudhakaran M, Doseff AI. The Targeted Impact of Flavones on Obesity-Induced Inflammation and the Potential Synergistic Role in Cancer and the Gut Microbiota. Molecules 2020; 25:E2477. [PMID: 32471061 PMCID: PMC7321129 DOI: 10.3390/molecules25112477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is an inflammatory disease that is approaching pandemic levels, affecting nearly 30% of the world's total population. Obesity increases the risk of diabetes, cardiovascular disorders, and cancer, consequentially impacting the quality of life and imposing a serious socioeconomic burden. Hence, reducing obesity and related life-threatening conditions has become a paramount health challenge. The chronic systemic inflammation characteristic of obesity promotes adipose tissue remodeling and metabolic changes. Macrophages, the major culprits in obesity-induced inflammation, contribute to sustaining a dysregulated immune function, which creates a vicious adipocyte-macrophage crosstalk, leading to insulin resistance and metabolic disorders. Therefore, targeting regulatory inflammatory pathways has attracted great attention to overcome obesity and its related conditions. However, the lack of clinical efficacy and the undesirable side-effects of available therapeutic options for obesity provide compelling reasons for the need to identify additional approaches for the prevention and treatment of obesity-induced inflammation. Plant-based active metabolites or nutraceuticals and diets with an increased content of these compounds are emerging as subjects of intense scientific investigation, due to their ability to ameliorate inflammatory conditions and offer safe and cost-effective opportunities to improve health. Flavones are a class of flavonoids with anti-obesogenic, anti-inflammatory and anti-carcinogenic properties. Preclinical studies have laid foundations by establishing the potential role of flavones in suppressing adipogenesis, inducing browning, modulating immune responses in the adipose tissues, and hindering obesity-induced inflammation. Nonetheless, the understanding of the molecular mechanisms responsible for the anti-obesogenic activity of flavones remains scarce and requires further investigations. This review recapitulates the molecular aspects of obesity-induced inflammation and the crosstalk between adipocytes and macrophages, while focusing on the current evidence on the health benefits of flavones against obesity and chronic inflammation, which has been positively correlated with an enhanced cancer incidence. We conclude the review by highlighting the areas of research warranting a deeper investigation, with an emphasis on flavones and their potential impact on the crosstalk between adipocytes, the immune system, the gut microbiome, and their role in the regulation of obesity.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Andrea I. Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
26
|
Liang Y, Liu D, Zhan J, Luo M, Han J, Wang P, Zhou Z. New insight into the mechanism of POP-induced obesity: Evidence from DDE-altered microbiota. CHEMOSPHERE 2020; 244:125123. [PMID: 32050320 DOI: 10.1016/j.chemosphere.2019.125123] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/27/2019] [Accepted: 10/13/2019] [Indexed: 06/10/2023]
Abstract
Although epidemiological studies demonstrate that persistent organic pollutants (POPs) could lead to metabolic syndrome, the mechanism has remained unclear. The dysbiosis of gut microbiota and the lipid metabolome have been put forward in the pathophysiology of metabolic syndrome. In this study, we used dichlorodiphenyldichloroethylene (DDE) as an example to study the effects of POP-impaired microbial composition and metabolome homeostasis on metabolic syndrome. The results showed that DDE exposure increased body weight and fat content and impaired glucose homeostasis. Further investigation revealed that DDE induced gut dysbiosis as indicated by an increased Firmicutes-to-Bacteroidetes ratio, which may impact energy harvest efficiency. Meanwhile, the plasma lipid metabolome profile was significantly altered by DDE. Furthermore, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, and triacylglycerol were identified as key metabolites affected by DDE treatment, and these altered lipid metabolites were highly correlated with changed microbiota composition. This study provides novel insight into the underlying mechanism of POP-induced obesity and diabetes, pointing to gut microbiota as one of the targets.
Collapse
Affiliation(s)
- Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China; College of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30, Xueyuan Road, Beijing, 100083, PR China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Mai Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jiajun Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China.
| |
Collapse
|
27
|
Dietary Control of Ganglioside Expression in Mammalian Tissues. Int J Mol Sci 2019; 21:ijms21010177. [PMID: 31887977 PMCID: PMC6981639 DOI: 10.3390/ijms21010177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/24/2019] [Accepted: 12/24/2019] [Indexed: 12/16/2022] Open
Abstract
Gangliosides are series of glycosphingolipids containing sialic acids in the oligosaccharide portion in mammalian cells. Gangliosides are a component of cellular membranes and play roles in modulating membrane function and the activity of membrane proteins. Abnormal expression and metabolism of gangliosides lead to the onset of several conditions in humans, such as neurologic diseases, diabetes, and cancer. A number of studies have been carried out to date to investigate the role of gangliosides in these diseases, and the effect of diet on tissue expression of gangliosides has recently become a topic of interest in this field. As gangliosides are degraded in the intestinal tract, ingested food-derived gangliosides are not directly absorbed into tissues in vivo, but the degradation products can be absorbed and affect ganglioside expression in the tissues. Recent studies have also shown that the expression of gangliosides in tissue cells can be indirectly induced by controlling the expression of ganglioside metabolism-related genes via the diet. These results indicate that dietary control can regulate the expression levels of gangliosides in tissues, which is expected to play a role in preventing and treating ganglioside-related diseases. This review introduces recent studies on the effect of diet on the expression of gangliosides in tissues, with a focus on our findings.
Collapse
|
28
|
Rice Bran Reduces Weight Gain and Modulates Lipid Metabolism in Rats with High-Energy-Diet-Induced Obesity. Nutrients 2019; 11:nu11092033. [PMID: 31480353 PMCID: PMC6769848 DOI: 10.3390/nu11092033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 02/05/2023] Open
Abstract
Obesity has become an epidemic worldwide. It is a complex metabolic disorder associated with many serious complications and high morbidity. Rice bran is a nutrient-dense by product of the rice milling process. Asia has the world’s highest rice production (90% of the world’s rice production); therefore, rice bran is inexpensive in Asian countries. Moreover, the high nutritional value of the rice bran suggests its potential as a food supplement promoting health improvements, such as enhancing brain function, lowering blood pressure, and regulating pancreatic secretion. The present study evaluated the anti-obesity effect of rice bran in rats with high-energy diet (HED)-induced obesity. Male Sprague–Dawley rats were randomly divided into one of five diet groups (n = 10 per group) and fed the following for eight weeks: Normal diet with vehicle treatment, HED with vehicle, rice bran-0.5X (RB-0.5X) (2% wt/wt rice bran), RB-1.0X (4% wt/wt rice bran), and RB-2.0X (8% wt/wt rice bran). Rice bran (RB-1.0X and RB-2.0X groups) markedly reduced obesity, including body weight and adipocyte size. In addition, treating rats with HED-induced obesity using rice bran significantly reduced the serum uric acid and glucose as well as the liver triglyceride (TG) and total cholesterol (TC). Furthermore, administration of an HED to obese rats significantly affected hepatic lipid homeostasis by increasing phosphotidylcholine (PC; 18:2/22:6), diacylglycerol (DG; 18:2/16:0), DG (18:2/18:1), DG (18:1/16:0), cholesteryl ester (CE; 20:5), CE (28:2), TG (18:0/16:0/18:3), and glycerol-1-2-hexadecanoate 3-octadecanoate. However, the rice bran treatment demonstrated an anti-adiposity effect by partially reducing the HED-induced DG (18:2/18:1) and TG (18:0/16:0/18:3) increases in obese rats. In conclusion, rice bran could act as an anti-obesity supplement in rats, as demonstrated by partially reducing the HED-induced DG and TG increases in obese rats, and thus limit the metabolic diseases associated with obesity and the accumulation of body fat and hepatic lipids in rats.
Collapse
|
29
|
Ruiz-Canizales J, Domínguez-Avila JA, Wall-Medrano A, Ayala-Zavala JF, González-Córdova AF, Vallejo-Córdoba B, Salazar-López NJ, González-Aguilar GA. Fiber and phenolic compounds contribution to the hepatoprotective effects of mango diets in rats fed high cholesterol/sodium cholate. Phytother Res 2019; 33:2996-3007. [PMID: 31418509 DOI: 10.1002/ptr.6479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The present study evaluated the contribution of mango fiber (MF) and mango phenolic compounds (MP) to the hepatoprotective effect of freeze-dried mango pulp (FDM) cultivar (cv.) "Ataulfo" diets in high cholesterol/sodium cholate (HCC)-fed rats. Male Wistar rats were fed with a HCC diet for 12 weeks, either untreated, or supplemented with MF, MP, FDM, or a control diet (no HCC; n = 6/group). All mango treatments significantly decreased hepatic cholesterol deposition and altered its fatty acid profile, whereas MF and MP mitigated adipose tissue hypertrophy. MF caused a lower level of proinflammatory cytokines (IL-1α/β, IFN-γ, TNF-α) whereas FDM increased the anti-inflammatory ones (IL-4, 6, 10). Mango treatments increased catalase (CAT) activity and its mRNA expression; superoxide dismutase (SOD) activity was normalized by MF and FDM, but its activity was unrelated to its hepatic mRNA expression. Changes in CAT and SOD mRNA expression were unrelated to altered Nrf2 mRNA expression. Higher hepatic PPARα and LXRα mRNA levels were found in MP and MF. We concluded that MF and MP are highly bioactive, according to the documented hepatoprotection in HCC-fed rats; their mechanism of action appears to be related to modulating cholesterol and fatty acid metabolism as well as to stimulating the endogenous antioxidant system.
Collapse
Affiliation(s)
- Jacqueline Ruiz-Canizales
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Abraham Wall-Medrano
- Departamento de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - J Fernando Ayala-Zavala
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Aarón F González-Córdova
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Belinda Vallejo-Córdoba
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Gustavo A González-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| |
Collapse
|
30
|
Li Y, Xu Y, Jadhav K, Zhu Y, Yin L, Zhang Y. Hepatic Forkhead Box Protein A3 Regulates ApoA-I (Apolipoprotein A-I) Expression, Cholesterol Efflux, and Atherogenesis. Arterioscler Thromb Vasc Biol 2019; 39:1574-1587. [PMID: 31291759 DOI: 10.1161/atvbaha.119.312610] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine the role of hepatic FOXA3 (forkhead box A3) in lipid metabolism and atherosclerosis. Approach and Results: Hepatic FOXA3 expression was reduced in diabetic or high fat diet-fed mice or patients with nonalcoholic steatohepatitis. We then used adenoviruses to overexpress or knock down hepatic FOXA3 expression. Overexpression of FOXA3 in the liver increased hepatic ApoA-I (apolipoprotein A-I) expression, plasma HDL-C (high-density lipoprotein cholesterol) level, macrophage cholesterol efflux, and macrophage reverse cholesterol transport. In contrast, knockdown of hepatic FOXA3 expression had opposite effects. We further showed that FOXA3 directly bound to the promoter of the Apoa1 gene to regulate its transcription. Finally, AAV8 (adeno-associated virus serotype 8)-mediated overexpression of human FOXA3 in the hepatocytes of Apoe-/- (apolipoprotein E-deficient) mice raised plasma HDL-C levels and significantly reduced atherosclerotic lesions. CONCLUSIONS Hepatocyte FOXA3 protects against atherosclerosis by inducing ApoA-I and macrophage reverse cholesterol transport.
Collapse
Affiliation(s)
- Yuanyuan Li
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yanyong Xu
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Kavita Jadhav
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yingdong Zhu
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Liya Yin
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yanqiao Zhang
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| |
Collapse
|
31
|
Jiménez-Rojo N, Riezman H. On the road to unraveling the molecular functions of ether lipids. FEBS Lett 2019; 593:2378-2389. [PMID: 31166014 DOI: 10.1002/1873-3468.13465] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
Ether lipids are glycerolipids further classified into alkyl-ether and alkenyl-ether (also termed plasmalogens) lipids. The two ether lipid subclasses share the first steps of their synthesis. However, alkyl-ether and alkenyl-ether lipids differ in their structure and physico-chemical properties (featuring different head groups) and, thus, probably in their functions. Ether lipids have intermittent distribution across the evolutionary tree and defects in their synthesis have been shown to perturb cellular homeostasis and lead to disease in humans. Here, we review their structure, their interactions with other lipids, and their potential roles in cellular functions, such as membrane homeostasis and membrane trafficking. Moreover, we discuss still unclear aspects of these lipids such as their subcellular distribution, and the need to unravel their molecular functions as well as how novel tools to study lipid biology will help clarify these aspects.
Collapse
Affiliation(s)
- Noemi Jiménez-Rojo
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Switzerland
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Switzerland
| |
Collapse
|
32
|
Heintz MM, Kumar R, Rutledge MM, Baldwin WS. Cyp2b-null male mice are susceptible to diet-induced obesity and perturbations in lipid homeostasis. J Nutr Biochem 2019; 70:125-137. [PMID: 31202118 DOI: 10.1016/j.jnutbio.2019.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Obesity is an endemic problem in the United States and elsewhere, and data indicate that in addition to overconsumption, exposure to specific chemicals enhances obesity. CYP2B metabolizes multiple endo- and xenobiotics, and recent data suggests that repression of Cyp2b activity increases dyslipidemia and age-onset obesity, especially in males. To investigate the role played by Cyp2b in lipid homeostasis and obesity, we treated wildtype and Cyp2b-null mice with a normal (ND) or 60% high-fat diet (HFD) for 10 weeks and determined metabolic and molecular changes. Male HFD-fed Cyp2b-null mice weigh 15% more than HFD-fed wildtype mice, primarily due to an increase in white adipose tissue (WAT); however, Cyp2b-null female mice did not demonstrate greater body mass or WAT. Serum parameters indicate increased ketosis, leptin and cholesterol in HFD-fed Cyp2b-null male mice compared to HFD-fed wildtype mice. Liver triglycerides and liver:serum triglyceride ratios were higher than their similarly treated wildtype counterparts in Cyp2b-null male mice, indicating a role for Cyp2b in fatty acid metabolism regardless of diet. Furthermore, RNAseq demonstrates that hepatic gene expression in ND-fed Cyp2b-null male mice is similar to HFD-fed WT male mice, suggestive of fatty liver disease progression and a role for Cyp2b in lipid homeostasis. Females did not show as demonstrative changes in liver health, and significantly fewer changes in gene expression, as well as gene expression associated with liver disease. Overall our data indicates that the repression or inhibition of CYP2B may exacerbate metabolic disorders and cause obesity by perturbing fatty acid metabolism, especially in males.
Collapse
Affiliation(s)
- Melissa M Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634
| | - Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC 29634
| | | | - William S Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634; Biological Sciences, Clemson University, Clemson, SC 29634.
| |
Collapse
|
33
|
Yu Z, Mao C, Fu X, Ma M. High Density Lipoprotein from Egg Yolk (EYHDL) Improves Dyslipidemia by Mediating Fatty Acids Metabolism in High Fat Diet-induced Obese Mice. Food Sci Anim Resour 2019; 39:179-196. [PMID: 31149661 PMCID: PMC6533406 DOI: 10.5851/kosfa.2018.e38] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022] Open
Abstract
We investigated the effect of high density lipoprotein from egg yolk (EYHDL) on
serum, hepatic and fecal lipid and fatty acids (FAs) levels and on gene
expression involved in FAs metabolism. Male KM mice were fed either normal diet
(ND; n=20), high fat diet (HFD; n=20), or high fat diet containing
EYHDL (EYHDL; 0.6 mg/g, every day by oral gavage, n=20) for 100 days. At
the end of the experiment, the effects of treatments on biochemical parameters,
FAs profiles and involved gene expression were analyzed. Our results revealed
that EYHDL markedly suppressed the body weight gain, accumulation of abdominal
fat tissues, serum concentrations of LDL-cholesterol (LDL-C) and triglycerides,
hepatic triglycerides and cholesterol accumulation, while increased serum
concentration of HDL-cholesterol (HDL-C). EYHDL intake also increased total
cholesterol (TC) excretions compared with HFD group. Moreover, it alleviated the
severity of fatty liver and improved glucose and insulin tolerance compared with
HFD. More importantly, EYHDL partially normalized FAs profiles in serum, liver
and fecaces and neutralized the HFD-induced upregulation of SREBP-1c, Acaca,
Fasn, GPAT and Scd1. In conclusion, our findings indicate that EYHDL may have
the potential to improve metabolic disturbances that occur in HFD mice and can
be considered as an appropriate dietary recommendation for the treatment of
metabolic syndrome (MetS).
Collapse
Affiliation(s)
- Zhihui Yu
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Changyi Mao
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xing Fu
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Meihu Ma
- National R&D Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| |
Collapse
|
34
|
Clugston RD, Gao MA, Blaner WS. The Hepatic Lipidome: A Gateway to Understanding the Pathogenes is of Alcohol-Induced Fatty Liver. Curr Mol Pharmacol 2019; 10:195-206. [PMID: 26278391 DOI: 10.2174/1874467208666150817111419] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022]
Abstract
Chronic alcohol consumption can lead to the development of alcoholic fatty liver disease. The underlying pathogenic mechanisms however, have not been fully elucidated. Here, we review the current state of the art regarding the application of lipidomics to study alcohol's effect on hepatic lipids. It is clear that alcohol has a profound effect on the hepatic lipidome, with documented changes in the major lipid categories (i.e. fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterol lipids and prenol lipids). Alcohol's most striking effect is the marked change in the hepatic fatty acyl pool. This effect includes increased levels of 18-carbon fatty acyl chains incorporated into multiple lipid species, as well as a general shift toward increased unsaturation of fatty acyl moieties. In addition to our literature review, we also make several recommendations to consider when designing lipidomic studies into alcohol's effects. These recommendations include integration of lipidomic data with other measures of lipid metabolism, inclusion of multiple experimental time points, and presentation of quantitative data. We believe rigorous analysis of the hepatic lipidome can yield new insight into the pathogenesis of alcohol-induced fatty liver. While the existing literature has been largely descriptive, the field is poised to apply lipidomics to yield a new level of understanding on alcohol's effects on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Robin D Clugston
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2H7. Canada
| | - Madeleine A Gao
- Department of Medicine, Columbia University, New York, NY, 10032. United States
| | - William S Blaner
- Department of Medicine, Columbia University, New York, NY, 10032. United States
| |
Collapse
|
35
|
Otto AC, Gan-Schreier H, Zhu X, Tuma-Kellner S, Staffer S, Ganzha A, Liebisch G, Chamulitrat W. Group VIA phospholipase A2 deficiency in mice chronically fed with high-fat-diet attenuates hepatic steatosis by correcting a defect of phospholipid remodeling. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:662-676. [PMID: 30735855 DOI: 10.1016/j.bbalip.2019.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022]
Abstract
A defect of hepatic remodeling of phospholipids (PL) is seen in non-alcoholic fatty liver disease and steatohepatitis (NASH) indicating pivotal role of PL metabolism in this disease. The deletion of group VIA calcium-independent phospholipase A2 (iPla2β) protects ob/ob mice from hepatic steatosis (BBAlip 1861, 2016, 440-461), however its role in high-fat diet (HFD)-induced NASH is still elusive. Here, wild-type and iPla2β-null mice were subjected to chronic feeding with HFD for 6 months. We showed that protection was observed in iPla2β-null mice with an attenuation of diet-induced body and liver-weight gains, liver enzymes, serum free fatty acids as well as hepatic TG and steatosis scores. iPla2β deficiency under HFD attenuated the levels of 1-stearoyl lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), and lysophosphatidylinositol (LPI) as well as elevation of hepatic arachidonate, arachidonate-containing cholesterol esters and prostaglandin E2. More importantly, this deficiency rescued a defect in PL remodeling and attenuated the ratio of saturated and unsaturated PL. The protection by iPla2β deficiency was not observed during short-term HFD feeding of 3 or 5 weeks which showed no PL remodeling defect. In addition to PC/PE, this deficiency reversed the suppression of PC/PI and PE/PI among monounsaturated PL. However, this deficiency did not modulate hepatic PL contents and PL ratios in ER fractions, ER stress, fibrosis, and inflammation markers. Hence, iPla2β inactivation protected mice against hepatic steatosis and obesity during chronic dietary NASH by correcting PL remodeling defect and PI composition relative to PC and PE.
Collapse
Affiliation(s)
- Ann-Christin Otto
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Xingya Zhu
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Alexandra Ganzha
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
36
|
Wang J, Zhang L, Xiao R, Li Y, Liao S, Zhang Z, Yang W, Liang B. Plasma lipidomic signatures of spontaneous obese rhesus monkeys. Lipids Health Dis 2019; 18:8. [PMID: 30621707 PMCID: PMC6323686 DOI: 10.1186/s12944-018-0952-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity plays crucial roles in the pathogenesis of metabolic diseases such as hyperlipidemia, nonalcoholic fatty liver disease (NAFLD), and type 2 diabetes (T2D). The underlying mechanisms linking obesity to metabolic diseases are still less understandable. METHODS Previously, we screened a group of spontaneously obese rhesus monkeys. Here, we performed a plasma lipidomic analysis of normal and obese monkeys using gas chromatography/mass spectroscopy (GC/MS) and ultra-high performance liquid chromatography/mass spectroscopy (UPLC/MS). RESULTS In total, 143 lipid species were identified, quantified, and classified into free fatty acids (FFA), phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidylserine (PS), phosphatidylglycerol (PG), lysophosphatidylcholine (LPC), lysophosphatidic acid (LPA), and sphingomyelin (SM). Data analysis showed that the obese monkeys had increased levels of fatty acids palmitoleic acid (C16:1) and arachidonic acid (C20:4), FFA especially palmitic acid (C16:0), as well as certain PC species and SM species. Surprisingly, the plasma level of LPA-C16:0 was approximately four-fold greater in the obese monkeys. Conversely, the levels of most PE species were obviously reduced in the obese monkeys. CONCLUSION Collectively, our work suggests that lipids such as FFA C16:0 and 16:0-LPA may be potential candidates for the diagnosis and study of obesity-related diseases.
Collapse
Affiliation(s)
- Junlong Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Ruyue Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Yunhai Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Shasha Liao
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Wenhui Yang
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Department of Geriatrics, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bin Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming, 650223, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
37
|
Liu F, Wang M, Wang Y, Cao Y, Sun Z, Chen M, Tian X, Wan J, Huang C. Metabonomics Study on the Hepatoprotective Effect of Panax notoginseng Leaf Saponins Using UPLC/Q-TOF-MS Analysis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:559-575. [DOI: 10.1142/s0192415x19500290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Alcohol liver disease is a major public health problem associated with lifestyle. Our recent study demonstrated that the roots of Panax notoginseng saponins (PNS) exert hepatoprotective effects against alcohol consumption. Considering that the leaves of Panax notoginseng saponins (LPNS) have similar chemical ingredients with PNS, increased attention should be given to the hepatoprotective effects of LPNS. In this study, a metabonomic approach based on ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-QTOF/MS) was developed to evaluate the hepatoprotective effect of LPNS on alcoholic fatty liver and elucidate the interaction mechanisms. Results showed that the ethanol-induced metabolic perturbations were restored after treatment with LPNS. Furthermore, 12 potential biomarkers (11 upregulated and 1 downregulated) were identified by V-plot and orthogonal partial least square discriminant analysis. Changes in the levels of these metabolites indicated that glycerophospholipid and fatty acid metabolism were disturbed in alcoholic fatty liver mouse. Our findings demonstrated that the UHPLC–QTOF/MS-based metabonomic method may provide a useful means for exploring biomarkers involved in alcoholic fatty liver and elucidating the therapeutic effects of LPNS. This work also showed that the metabonomic approach is a powerful and promising tool for the evaluation of the efficacy of traditional Chinese medicine and elucidation of related mechanisms.
Collapse
Affiliation(s)
- Fang Liu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai Zhangjiang Hitech Park, Shanghai 201203, P. R. China
| | - Meng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Yang Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Yiwei Cao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Zhaolin Sun
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai Zhangjiang Hitech Park, Shanghai 201203, P. R. China
| | - Mingcang Chen
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai Zhangjiang Hitech Park, Shanghai 201203, P. R. China
| | - Xiaoting Tian
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai Zhangjiang Hitech Park, Shanghai 201203, P. R. China
| | - Jianbo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Chenggang Huang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai Zhangjiang Hitech Park, Shanghai 201203, P. R. China
| |
Collapse
|
38
|
Ma HF, Wei F, Wu BF, Yang C, Xie Y, Wu ZY, Lv X, Chen H. Profiling and quantification of aminophospholipids based on chemical derivatization coupled with HPLC-MS. J Lipid Res 2018; 60:121-134. [PMID: 30482807 DOI: 10.1194/jlr.m089482] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/01/2018] [Indexed: 01/21/2023] Open
Abstract
In this study, a novel strategy based on acetone stable-isotope derivatization coupled with HPLC-MS for profiling and accurate quantification of aminophospholipids (phosphatidylethanolamine and phosphatidylserine) in biological samples was developed. Acetone derivatization leads to alkylation of the primary amino groups of aminophospholipids with an isopropyl moiety; the use of deuterium-labeled acetone (d6-acetone) introduced a 6 Da mass shift that was ideally suited for profiling and quantification analysis with high selectivity and accuracy. After derivatization, significantly increased column efficiency for chromatographic separation and detection sensitivity for MS analysis of aminophospholipids was observed. Furthermore, an accuracy quantification method was developed. Aminophospholipids in biological samples were derivatized with d0-acetone; while more than two aminophospholipid standards were selected for each class of aminophospholipid and derivatized with d6-acetone, which were then used as the internal standards to typically construct a calibration curve for each class to normalize the nonuniformity response caused by the differential fragmentation kinetics resulting from the distinct chemical constitution of individual aminophospholipid species in the biological samples. The excellent applicability of the developed method was validated by profiling and quantification of aminophospholipids presented in liver samples from rats fed with different diets.
Collapse
Affiliation(s)
- Hui-Fang Ma
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Fang Wei
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Bang-Fu Wu
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Chen Yang
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Ya Xie
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Zong-Yuan Wu
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Xin Lv
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Hong Chen
- Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Key Laboratory of Oilseeds Processing of Ministry of Agriculture, Key Laboratory of Biology and Genetic Improvement of Oil Crops of Ministry of Agriculture, and Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| |
Collapse
|
39
|
Just S, Mondot S, Ecker J, Wegner K, Rath E, Gau L, Streidl T, Hery-Arnaud G, Schmidt S, Lesker TR, Bieth V, Dunkel A, Strowig T, Hofmann T, Haller D, Liebisch G, Gérard P, Rohn S, Lepage P, Clavel T. The gut microbiota drives the impact of bile acids and fat source in diet on mouse metabolism. MICROBIOME 2018; 6:134. [PMID: 30071904 PMCID: PMC6091023 DOI: 10.1186/s40168-018-0510-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/02/2018] [Indexed: 05/06/2023]
Abstract
BACKGROUND As the gut microbiota contributes to metabolic health, it is important to determine specific diet-microbiota interactions that influence host metabolism. Bile acids and dietary fat source can alter phenotypes of diet-induced obesity, but the interplay with intestinal microorganisms is unclear. Here, we investigated metabolic consequences of diets enriched in primary bile acids with or without addition of lard or palm oil, and studied gut microbiota structure and functions in mice. RESULTS In combination with bile acids, dietary lard fed to male C57BL/6N mice for a period of 8 weeks enhanced fat mass accumulation in colonized, but not in germ-free mice when compared to palm oil. This was associated with impaired glucose tolerance, lower fasting insulin levels, lower counts of enteroendocrine cells, fatty liver, and elevated amounts of hepatic triglycerides, cholesteryl esters, and monounsaturated fatty acids. Lard- and bile acid-fed mice were characterized by shifts in dominant gut bacterial communities, including decreased relative abundances of Lachnospiraceae and increased occurrence of Desulfovibrionaceae and the species Clostridium lactatifermentans and Flintibacter butyricus. Metatranscriptomic analysis revealed shifts in microbial functions, including lipid and amino acid metabolism. CONCLUSIONS Caution is required when interpreting data from diet-induced obesity models due to varying effects of dietary fat source. Detrimental metabolic consequences of a diet enriched with lard and primary bile acids were dependent on microbial colonization of the host and were linked to hepatic lipid rearrangements and to alterations of dominant bacterial communities in the cecum.
Collapse
Affiliation(s)
- Sarah Just
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Stanislas Mondot
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Josef Ecker
- Nutritional Physiology, Technical University of Munich, Freising, Germany
| | - Katrin Wegner
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Eva Rath
- Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Laura Gau
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Theresa Streidl
- Institute of Medical Microbiology, Functional Microbiome Research Group, University Hospital of RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Genevieve Hery-Arnaud
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Sinah Schmidt
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Till Robin Lesker
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Valentin Bieth
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Andreas Dunkel
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Till Strowig
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Hofmann
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Dirk Haller
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Sascha Rohn
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Thomas Clavel
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
- Institute of Medical Microbiology, Functional Microbiome Research Group, University Hospital of RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
40
|
Eisinger K, Rein-Fischboeck L, Neumeier M, Schmidhofer S, Pohl R, Haberl EM, Liebisch G, Kopp A, Schmid A, Krautbauer S, Buechler C. Alpha-syntrophin deficient mice are protected from adipocyte hypertrophy and ectopic triglyceride deposition in obesity. Exp Mol Pathol 2018; 104:212-221. [DOI: 10.1016/j.yexmp.2018.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
|
41
|
de Visser HM, Mastbergen SC, Ravipati S, Welsing PMJ, Pinto FC, Lafeber FPJG, Chapman V, Barrett DA, Weinans H. Local and systemic inflammatory lipid profiling in a rat model of osteoarthritis with metabolic dysregulation. PLoS One 2018; 13:e0196308. [PMID: 29684084 PMCID: PMC5912715 DOI: 10.1371/journal.pone.0196308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/10/2018] [Indexed: 01/15/2023] Open
Abstract
Objective Bioactive oxidised lipids (oxylipins) are important signalling mediators, capable of modulating the inflammatory state of the joint and anticipated to be of importance in joint homeostasis and status of osteoarthritis. The aim of this study was to quantify oxylipin levels in plasma and synovial fluid from rats with experimentally induced osteoarthritis to investigate the potential role of oxylipins as a marker in the disease process of early osteoarthritis. Design Forty rats were randomly allocated to a standard or high-fat diet group. After 12 weeks, local cartilage damage was induced in one knee joint in 14 rats of each diet group. The remaining 6 rats per group served as controls. At week 24, samples were collected. Oxylipin levels were quantified by liquid chromatography–mass spectrometry. Results Overall, 31 lipid-derived inflammatory mediators were detected in fasted plasma and synovial fluid. Principal component analysis identified four distinct clusters associated with histopathological changes. Diet induced differences were evident for 13 individual plasma oxylipins, as well as 5,6-EET in synovial fluid. Surgical-model induced differences were evident for three oxylipins in synovial fluid (15-HETE, 8,9-DHET and 17R-ResolvinD1) with a different response in lipid concentrations for synovial fluid and plasma. Conclusions We demonstrate the quantification of oxidised lipids in rat plasma and synovial fluid in a model of early experimental osteoarthritis. Oxylipins in the synovial fluid that were altered as consequence of the surgically induced osteoarthritis were not represented in the plasma. Our findings suggest differential roles of the oxylipins in the local versus peripheral compartment.
Collapse
Affiliation(s)
- H. M. de Visser
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S. C. Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| | - S. Ravipati
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - P. M. J. Welsing
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. C. Pinto
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - F. P. J. G. Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - V. Chapman
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
- Arthritis Research UK Pain Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kindom
| | - D. A. Barrett
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - H. Weinans
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
42
|
Rein-Fischboeck L, Haberl EM, Pohl R, Schmid V, Feder S, Krautbauer S, Liebisch G, Buechler C. Alpha-syntrophin null mice are protected from non-alcoholic steatohepatitis in the methionine-choline-deficient diet model but not the atherogenic diet model. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:526-537. [PMID: 29474931 DOI: 10.1016/j.bbalip.2018.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/23/2018] [Accepted: 02/17/2018] [Indexed: 12/13/2022]
Abstract
Adipose tissue dysfunction contributes to the pathogenesis of non-alcoholic steatohepatitis (NASH). The adapter protein alpha-syntrophin (SNTA) is expressed in adipocytes. Knock-down of SNTA increases preadipocyte proliferation and formation of small lipid droplets, which are both characteristics of healthy adipose tissue. To elucidate a potential protective role of SNTA in NASH, SNTA null mice were fed a methionine-choline-deficient (MCD) diet or an atherogenic diet which are widely used as preclinical NASH models. MCD diet mediated loss of fat mass was largely improved in SNTA-/- mice compared to the respective wild type animals. Hepatic lipids were mostly unchanged while the oxidative stress marker malondialdehyde was only induced in the wild type mice. The expression of inflammatory markers and macrophage immigration into the liver were reduced in SNTA-/- animals. This protective function of SNTA loss was absent in atherogenic diet induced NASH. Here, hepatic expression of inflammatory and fibrotic genes was similar in both genotypes though mutant mice gained less body fat during feeding. Hepatic cholesterol and ceramide were strongly induced in both strains upon feeding the atherogenic diet, while hepatic sphingomyelin, phosphatidylserine and phosphatidylethanolamine levels were suppressed. SNTA deficient mice are protected from fat loss and NASH in the experimental MCD model. NASH induced by an atherogenic diet is not influenced by loss of SNTA. The present study suggests the use of different experimental NASH models to study the pathophysiological role of proteins like SNTA in NASH.
Collapse
Affiliation(s)
- Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Verena Schmid
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Susanne Feder
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany.
| |
Collapse
|
43
|
Abstract
Ether lipids, such as plasmalogens, are peroxisome-derived glycerophospholipids in which the hydrocarbon chain at the sn-1 position of the glycerol backbone is attached by an ether bond, as opposed to an ester bond in the more common diacyl phospholipids. This seemingly simple biochemical change has profound structural and functional implications. Notably, the tendency of ether lipids to form non-lamellar inverted hexagonal structures in model membranes suggests that they have a role in facilitating membrane fusion processes. Ether lipids are also important for the organization and stability of lipid raft microdomains, cholesterol-rich membrane regions involved in cellular signaling. In addition to their structural roles, a subset of ether lipids are thought to function as endogenous antioxidants, and emerging studies suggest that they are involved in cell differentiation and signaling pathways. Here, we review the biology of ether lipids and their potential significance in human disorders, including neurological diseases, cancer, and metabolic disorders.
Collapse
Affiliation(s)
- John M Dean
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
44
|
Dagla I, Benaki D, Baira E, Lemonakis N, Poudyal H, Brown L, Tsarbopoulos A, Skaltsounis AL, Mikros E, Gikas E. Alteration in the liver metabolome of rats with metabolic syndrome after treatment with Hydroxytyrosol. A Mass Spectrometry And Nuclear Magnetic Resonance - based metabolomics study. Talanta 2018; 178:246-257. [DOI: 10.1016/j.talanta.2017.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/04/2017] [Accepted: 09/10/2017] [Indexed: 01/14/2023]
|
45
|
Hu L, Bo L, Zhang M, Li S, Zhao X, Sun C. Metabonomics analysis of serum from rats given long-term and low-level cadmium by ultra-performance liquid chromatography-mass spectrometry. Xenobiotica 2017; 48:1079-1088. [PMID: 29143552 DOI: 10.1080/00498254.2017.1397811] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
1. This study evaluated the toxicity of chronic exposure to low-level cadmium (Cd) in rats using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). Forty male Sprague-Dawley rats were randomly assigned to four groups, namely, the control group, low-dose group (0.13 mg/kg·bw), middle-dose group (0.8 mg/kg·bw) and high-dose group (4.89 mg/kg·bw). The rats continuously received CdCl2 via drinking water for 24 weeks. Serum samples were collected for metabonomics analysis. The data generated from the UPLC-MS was analysed using principal components analysis (PCA) and partial least-squares discriminant analysis (PLS-DA). PLS-DA model with satisfactory explanatory and predictive ability is capable of discriminating the treatment groups from the control group. 2. Finally, the 10 metabolites were identified and showed significant changes in some treatment groups compared with that in the control group (p < 0.0167 or p < 0.003). Exposure to Cd resulted in increased intensities of lysophosphatidic acid (P-16:0e/0:0), glycocholic acid, bicyclo-prostaglandin E2, lithocholyltaurine, sulfolithocholylglycine, lysophosphatidylethanolamine (20:5/0:0) and lysophosphatidylcholine (20:0), as well as decreased intensities of 3-indolepropionic acid, phosphatidylcholine (18:4/18:0) and 15S-hydroxyeicosatrienoic acid in rat serum. 3. Results suggest that exposure to Cd can cause disturbances in the lipid metabolism, amino acid metabolism, nervous system, antioxidant defence system, liver and kidney function.
Collapse
Affiliation(s)
- Liyan Hu
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| | - Lu Bo
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| | - Meiyan Zhang
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| | - Siqi Li
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| | - Xiujuan Zhao
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| | - Changhao Sun
- a Department of Nutrition and Food Hygiene , Public Health College, Harbin Medical University , Harbin , China
| |
Collapse
|
46
|
Pati S, Krishna S, Lee JH, Ross MK, de La Serre CB, Harn DA, Wagner JJ, Filipov NM, Cummings BS. Effects of high-fat diet and age on the blood lipidome and circulating endocannabinoids of female C57BL/6 mice. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:26-39. [PMID: 28986283 DOI: 10.1016/j.bbalip.2017.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/17/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
Alterations in lipid metabolism play a significant role in the pathogenesis of obesity-associated disorders, and dysregulation of the lipidome across multiple diseases has prompted research to identify novel lipids indicative of disease progression. To address the significant gap in knowledge regarding the effect of age and diet on the blood lipidome, we used shotgun lipidomics with electrospray ionization-mass spectrometry (ESI-MS). We analyzed blood lipid profiles of female C57BL/6 mice following high-fat diet (HFD) and low-fat diet (LFD) consumption for short (6weeks), long (22weeks), and prolonged (36weeks) periods. We examined endocannabinoid levels, plasma esterase activity, liver homeostasis, and indices of glucose tolerance and insulin sensitivity to compare lipid alterations with metabolic dysregulation. Multivariate analysis indicated differences in dietary blood lipid profiles with the most notable differences after 6weeks along with robust alterations due to age. HFD altered phospholipids, fatty acyls, and glycerolipids. Endocannabinoid levels were affected in an age-dependent manner, while HFD increased plasma esterase activity at all time points, with the most pronounced effect at 6weeks. HFD-consumption also altered liver mRNA levels of PPARα, PPARγ, and CD36. These findings indicate an interaction between dietary fat consumption and aging with widespread effects on the lipidome, which may provide a basis for identification of female-specific obesity- and age-related lipid biomarkers.
Collapse
Affiliation(s)
- Sumitra Pati
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States
| | - Saritha Krishna
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| | - Jung Hwa Lee
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39759, United States
| | - Matthew K Ross
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39759, United States
| | - Claire B de La Serre
- Department of Foods and Nutrition, College of Family and Consumer Sciences, University of Georgia, Athens, GA 30602, United States
| | - Donald A Harn
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States
| | - John J Wagner
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States; Interdisciplinary Toxicology Program, University of, Georgia, Athens, GA 30602, United States
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, United States; Interdisciplinary Toxicology Program, University of, Georgia, Athens, GA 30602, United States.
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States; Interdisciplinary Toxicology Program, University of, Georgia, Athens, GA 30602, United States.
| |
Collapse
|
47
|
Weiss TS, Lupke M, Ibrahim S, Buechler C, Lorenz J, Ruemmele P, Hofmann U, Melter M, Dayoub R. Attenuated lipotoxicity and apoptosis is linked to exogenous and endogenous augmenter of liver regeneration by different pathways. PLoS One 2017; 12:e0184282. [PMID: 28877220 PMCID: PMC5587239 DOI: 10.1371/journal.pone.0184282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) covers a spectrum from simple steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. Free fatty acids (FFA) induce steatosis and lipo-toxicity and correlate with severity of NAFLD. In this study we aimed to investigate the role of exogenous and endogenous ALR (augmenter of liver regeneration) for FFA induced ER (endoplasmatic reticulum) -stress and lipoapoptosis. Primary human hepatocytes or hepatoma cells either treated with recombinant human ALR (rhALR, 15kDa) or expressing short form ALR (sfALR, 15kDa) were incubated with palmitic acid (PA) and analyzed for lipo-toxicity, -apoptosis, activation of ER-stress response pathways, triacylglycerides (TAG), mRNA and protein expression of lipid metabolizing genes. Both, exogenous rhALR and cytosolic sfALR reduced PA induced caspase 3 activity and Bax protein expression and therefore lipotoxicity. Endogenous sfALR but not rhALR treatment lowered TAG levels, diminished activation of ER-stress mediators C-Jun N-terminal kinase (JNK), X-box binding protein-1 (XBP1) and proapoptotic transcription factor C/EBP-homologous protein (CHOP), and reduced death receptor 5 protein expression. Cellular ALR exerts its lipid lowering and anti-apoptotic actions by enhancing FABP1, which binds toxic FFA, increasing mitochondrial β-oxidation by elevating the mitochondrial FFA transporter CPT1α, and decreasing ELOVL6, which delivers toxic FFA metabolites. We found reduced hepatic mRNA levels of ALR in a high fat diet mouse model, and of ALR and FOXA2, a transcription factor inducing ALR expression, in human steatotic as well as NASH liver samples, which may explain increased lipid deposition and reduced β-oxidation in NASH patients. Present study shows that exogenous and endogenous ALR reduce PA induced lipoapoptosis. Furthermore, cytosolic sfALR changes mRNA and protein expression of genes regulating lipid metabolism, reduces ER-stress finally impeding progression of NASH.
Collapse
Affiliation(s)
- Thomas S. Weiss
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
- Center for Liver Cell Research, University of Regensburg Hospital, Regensburg, Germany
- * E-mail:
| | - Madeleine Lupke
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
| | - Sara Ibrahim
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Julia Lorenz
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
| | - Petra Ruemmele
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Germany
| | - Michael Melter
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
| | - Rania Dayoub
- Children’s University Hospital, University of Regensburg, Regensburg, Germany
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
| |
Collapse
|
48
|
Jurowski K, Kochan K, Walczak J, Barańska M, Piekoszewski W, Buszewski B. Analytical Techniques in Lipidomics: State of the Art. Crit Rev Anal Chem 2017; 47:418-437. [PMID: 28340309 DOI: 10.1080/10408347.2017.1310613] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Current studies related to lipid identification and determination, or lipidomics in biological samples, are one of the most important issues in modern bioanalytical chemistry. There are many articles dedicated to specific analytical strategies used in lipidomics in various kinds of biological samples. However, in such literature, there is a lack of articles dedicated to a comprehensive review of the actual analytical methodologies used in lipidomics. The aim of this article is to characterize the lipidomics methods used in modern bioanalysis according to the methodological point of view: (1) chromatography/separation methods, (2) spectroscopic methods and (3) mass spectrometry and also hyphenated methods. In the first part, we discussed thin layer chromatography (TLC), high-pressure liquid chromatography (HPLC), gas chromatography (GC) and capillary electrophoresis (CE). The second part includes spectroscopic techniques such as Raman spectroscopy (RS), Fourier transform infrared spectroscopy (FT-IR) and nuclear magnetic resonance (NMR). The third part is a synthetic review of mass spectrometry, matrix-assisted laser desorption/ionization (MALDI), hyphenated methods, which include liquid chromatography-mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS) and also multidimensional techniques. Other aspects are the possibilities of the application of the described methods in lipidomics studies. Due to the fact that the exploration of new methods of lipidomics analysis and their applications in clinical and medical studies are still challenging for researchers working in life science, we hope that this review article will be very useful for readers.
Collapse
Affiliation(s)
- Kamil Jurowski
- a Kraków Higher School of Health Promotion , Krakow , Poland
| | - Kamila Kochan
- b Jagiellonian Centre for Experimental Therapeutics (JCET) , Jagiellonian University in Cracow , Cracow , Poland.,c Centre for Biospectroscopy and School of Chemistry , Monash University , Clayton , Victoria , Australia
| | - Justyna Walczak
- d Department of Environmental Chemistry and Bioanalytics , Faculty of Chemistry, Nicolaus Copernicus University , Torun , Poland
| | - Małgorzata Barańska
- b Jagiellonian Centre for Experimental Therapeutics (JCET) , Jagiellonian University in Cracow , Cracow , Poland.,e Department of Chemical Physics, Faculty of Chemistry , Jagiellonian University in Cracow , Cracow , Poland
| | - Wojciech Piekoszewski
- f Department of Analytical Chemistry, Faculty of Chemistry , Jagiellonian University in Cracow , Cracow , Poland.,g School of Biomedicine , Far Eastern Federal University , Vladivostok , Russia
| | - Bogusław Buszewski
- d Department of Environmental Chemistry and Bioanalytics , Faculty of Chemistry, Nicolaus Copernicus University , Torun , Poland
| |
Collapse
|
49
|
Park HM, Park KT, Park EC, Kim SI, Choi MS, Liu KH, Lee CH. Mass Spectrometry-Based Metabolomic and Lipidomic Analyses of the Effects of Dietary Platycodon grandiflorum on Liver and Serum of Obese Mice under a High-Fat Diet. Nutrients 2017; 9:nu9010071. [PMID: 28106735 PMCID: PMC5295115 DOI: 10.3390/nu9010071] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/27/2016] [Accepted: 01/07/2017] [Indexed: 12/23/2022] Open
Abstract
We aimed to identify metabolites involved in the anti-obesity effects of Platycodon grandiflorum (PG) in high-fat diet (HFD)-fed mice using mass spectrometry (MS)-based metabolomic techniques. C57BL/6J mice were divided into four groups: normal diet (ND)-fed mice, HFD-fed mice, HFD with 1% PG extract-fed mice (HPGL), and HFD with 5% PG extract-fed mice (HPGH). After 8 weeks, the HFD group gained more weight than the ND group, while dietary 5% PG extract attenuated this change. The partial least squares discriminant analysis (PLS-DA) score plots showed a clear distinction between experimental groups in serum and liver markers. We also identified 10 and 32 metabolites in the serum and liver, respectively, as potential biomarkers that could explain the effect of high-dose PG added to HFD-fed mice, which were strongly involved in amino acid metabolism (glycine, serine, threonine, methionine, glutamate, phenylalanine, ornithine, lysine, and tyrosine), TCA cycle (fumarate and succinate), lipid metabolism (linoleic and oleic acid methyl esters, oleamide, and cholesterol), purine/pyrimidine metabolism (uracil and hypoxanthine), carbohydrate metabolism (maltose), and glycerophospholipid metabolism (phosphatidylcholines, phosphatidylethanolamines, lysophosphatidylcholines, and lysophosphatidylethanolamines). We suggest that further studies on these metabolites could help us gain a better understanding of both HFD-induced obesity and the effects of PG.
Collapse
Affiliation(s)
- Hye Min Park
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Kab-Tae Park
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Edmond Changkyun Park
- Division of Life Science, Korea Basic Science Institute, Daejeon 34133, Korea.
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Seung Ii Kim
- Division of Life Science, Korea Basic Science Institute, Daejeon 34133, Korea.
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Myung Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Korea.
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
50
|
Tubulin alpha 8 is expressed in hepatic stellate cells and is induced in transformed hepatocytes. Mol Cell Biochem 2017; 428:161-170. [DOI: 10.1007/s11010-016-2926-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022]
|