1
|
Kang JH, Toita R, Kawano T, Murata M, Kano A. Phospholipids and their metabolites as diagnostic biomarkers of human diseases. Prog Lipid Res 2025; 99:101340. [PMID: 40409729 DOI: 10.1016/j.plipres.2025.101340] [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: 01/16/2025] [Revised: 04/03/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Phospholipids that occur predominantly in the plasma membrane of mammalian cells are phosphatidylcholine (PC), phosphatidylserine (PS), phosphatidylethanolamine (PE), sphingomyelin (SM), and phosphatidylinositol (or phosphoinositide; PI). These membrane phospholipids are a promising source of disease-related biomarkers. Phospholipids and their metabolites are altered by the type of disease or disease progression. Metabolomics has shown that increased or decreased levels of altered phospholipids and their metabolites can be useful indicators for the diagnosis of various human diseases. In this review, we discuss the utility of the five major membrane phospholipids (PC, PS, PE, and SM, and PI) and their metabolites as diagnostic biomarkers of human diseases.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Arihiro Kano
- Institute for Materials Chemistry and Engineering (IMCE), Kyushu University, 6-1 Kasuga-kouen, Kasuga, Fukuoka 819-0395, Japan
| |
Collapse
|
2
|
Zhang R, Li Z, Huang L, Kong W, Zheng Y, Wang Y, Shen X, Huang L, Wang X, Zheng Q, Wu L, Ke Y, Mao R, Peng Z, Sun C, Feng ST, Lin S, Wang Y, Li X. Altered gut microbiome-metabolite interactions link intestinal inflammation severity and MR enterography abnormalities in Crohn's disease. iScience 2025; 28:112310. [PMID: 40292324 PMCID: PMC12033948 DOI: 10.1016/j.isci.2025.112310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/06/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Altered gut microbiota‒metabolite interactions may result in intestinal inflammation severity variation in Crohn's disease (CD). Magnetic resonance enterography (MRE) advances anti-inflammatory strategy development. We aimed to identify inflammation-related multiomics factors and MRE interactions for CD management, analyzing 425 CD patients and 42 healthy controls undergoing MRE, ileocolonoscopy, and fecal/blood sampling (microbiota/metabolite analyses), with intestinal inflammation categorized by MRE and ileocolonoscopy. Ruminococcus species were enriched in CD patients versus healthy controls, while Pseudomonas and Staphylococcus dominated moderate-severe versus no-mild inflammation groups, suggesting inflammation-level associations. Ruminococcus gauvreauii suppressed intestinal inflammation by regulating serum PC(O-34:3), ePE(38:6), and ceramides (all p < 0.05). Serum N-acetylneuraminic acid and guanidinoacetic acid correlated with intestinal morphological changes (e.g., MRE-detectable effusion and wall thickness) and inflammation severity (P ACME < 0.05). A link was established between microscopic microbiota-metabolite markers and macroscopic imaging of inflammatory features, which could offer valuable insights into inflammation management.
Collapse
Affiliation(s)
- Ruonan Zhang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Zhoulei Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Li Huang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Weimiao Kong
- Youth Innovation Team of Medical Bioinformatics, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Cell Biology and Genetics, College of Basic Medicine, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yidong Zheng
- Youth Innovation Team of Medical Bioinformatics, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Cell Biology and Genetics, College of Basic Medicine, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yangdi Wang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Xiaodi Shen
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Lili Huang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Xinyue Wang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Qingzhu Zheng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Luyao Wu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Yaoqi Ke
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, P.R. China
| | - Zhenpeng Peng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Canhui Sun
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Shaochun Lin
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| | - Yejun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, P.R. China
| | - Xuehua Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan II Road, Guangzhou 510080, People's Republic of China
| |
Collapse
|
3
|
Huang M, Zhang Y, Xu X, Duan R, Yang H. Chronic chlorothalonil exposure inhibits locomotion and interferes with the gut-liver axis in Pelophylax nigromaculatus tadpoles. Sci Rep 2025; 15:14573. [PMID: 40280937 PMCID: PMC12032272 DOI: 10.1038/s41598-025-98081-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Chlorothalonil is a widely used fungicide that has a negative effect on individual movement, but its impact pathway needs further refinement. Here, the effects of exposure to chlorothalonil on the locomotion behavior of Pelophylax nigromaculatus tadpoles (GS23) were measured at three different levels (0 µg/L, 10 µg/L, and 50 µg/L), and the possible pathways of its effects were analyzed from the gut-liver axis. Chlorothalonil exposure levels of 10 µg/L and 50 µg/L significantly reduced the average speed of P. nigromaculatus tadpoles by 26% and 32.7%, respectively, and significantly decreased the locomotor frequency by 27.1% and 58.6%, respectively. Gut microbiota analysis revealed chlorothalonil exposure significantly increased the abundance of Firmicutes, while significantly decreased the abundance of Actinobacteriota, Pseudomonas, and Rhodococcus. Metabolomics analysis identified that chlorothalonil treatment changed amino acid-related metabolism pathways in the gut and liver and altered the glycerophospholipid metabolism pathway in the liver. This study indicated that chlorothalonil can affect individual locomotor abilities and interfering with the gut-liver axis of aquatic animals. These findings establish that chlorothalonil compromises aquatic organism motility through a multi-target mechanism involving gut microbiota modulation, amino acid metabolic interference, and hepatic lipid pathway disruption.
Collapse
Affiliation(s)
- Minyi Huang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
- Key Laboratory of Development, Utilization, Quality and Safety Control of Characteristic Agricultural Resources in Central Hunan Province, Loudi, 417000, Hunan, China
| | - Yuhao Zhang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| | - Xiang Xu
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| | - Renyan Duan
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China.
- Key Laboratory of Development, Utilization, Quality and Safety Control of Characteristic Agricultural Resources in Central Hunan Province, Loudi, 417000, Hunan, China.
| | - Hui Yang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| |
Collapse
|
4
|
Vermeulen I, Li M, van Mourik H, Yadati T, Eijkel G, Balluff B, Godschalk R, Temmerman L, Biessen EAL, Kulkarni A, Theys J, Houben T, Cillero‐Pastor B, Shiri‐Sverdlov R. Inhibition of intracellular versus extracellular cathepsin D differentially alters the liver lipidome of mice with metabolic dysfunction-associated steatohepatitis. FEBS J 2025; 292:1781-1797. [PMID: 39726152 PMCID: PMC11970712 DOI: 10.1111/febs.17358] [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: 03/14/2024] [Revised: 08/09/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) progressing to metabolic dysfunction-associated steatohepatitis (MASH), characterized by hepatic inflammation, has significantly increased in recent years due to unhealthy dietary practices and sedentary lifestyles. Cathepsin D (CTSD), a lysosomal protease involved in lipid homeostasis, is linked to abnormal lipid metabolism and inflammation in MASH. Although primarily intracellular, CTSD can be secreted extracellularly. Our previous proteomics research has shown that inhibition of extracellular CTSD results in more anti-inflammatory effects and fewer potential side effects compared to intracellular CTSD inhibition. However, the correlation between reduced side effects and alterations in the hepatic lipid composition remains unknown. This study aims to investigate the correlation between intra- and extracellular CTSD inhibition and potential alterations in the hepatic lipid composition in MASH. Low-density lipoprotein receptor knockout (Ldlr-/-) mice were fed a high-fat diet for 10 weeks and received subcutaneous injections every 2 days of vehicle, intracellular CTSD inhibitor (GA-12), or extracellular CTSD inhibitor (CTD-002). Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) was used to visualize and compare the lipid composition in liver tissues. Hepatic phosphatidylcholine remodeling was observed with both inhibitors, suggesting their therapeutic potential in treating MASH. Treatment with an intracellular CTSD inhibitor resulted in elevated levels of cardiolipin, reactive oxygen species, phosphatidylinositol, phosphatidylethanolamine, and lipids that are linked to mitochondrial dysfunction and inflammation, and induced more oxidative stress. The observed modifications in lipid composition demonstrate the clinical advantages of extracellular CTSD inhibition as a potentially beneficial therapeutic approach for MASH.
Collapse
Affiliation(s)
- Isabeau Vermeulen
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Mengying Li
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Hester van Mourik
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
- Department of Precision Medicine, Institute for Oncology and Reproduction (GROW)Maastricht UniversityThe Netherlands
| | - Tulasi Yadati
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Gert Eijkel
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Benjamin Balluff
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
| | - Roger Godschalk
- Department of Pharmacology and Toxicology, Institute for Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical Center (UMC)The Netherlands
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical Center (UMC)The Netherlands
- Institute for Molecular Cardiovascular ResearchRheinisch‐Westfälische Technische Hochschule (RWTH) Aachen UniversityGermany
| | | | - Jan Theys
- Department of Precision Medicine, Institute for Oncology and Reproduction (GROW)Maastricht UniversityThe Netherlands
| | - Tom Houben
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| | - Berta Cillero‐Pastor
- Maastricht Multimodal Molecular Imaging Institute (M4i)University of MaastrichtThe Netherlands
- Cell Biology‐Inspired Tissue Engineering (cBITE), MERLNMaastricht UniversityThe Netherlands
| | - Ronit Shiri‐Sverdlov
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityThe Netherlands
| |
Collapse
|
5
|
Queathem ED, Moazzami Z, Stagg DB, Nelson AB, Fulghum K, Hayir A, Seay A, Gillingham JR, d’Avignon DA, Han X, Ruan HB, Crawford PA, Puchalska P. Ketogenesis supports hepatic polyunsaturated fatty acid homeostasis via fatty acid elongation. SCIENCE ADVANCES 2025; 11:eads0535. [PMID: 39879309 PMCID: PMC11777252 DOI: 10.1126/sciadv.ads0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Ketogenesis is a dynamic metabolic conduit supporting hepatic fat oxidation particularly when carbohydrates are in short supply. Ketone bodies may be recycled into anabolic substrates, but a physiological role for this process has not been identified. Here, we use mass spectrometry-based 13C-isotope tracing and shotgun lipidomics to establish a link between hepatic ketogenesis and lipid anabolism. Unexpectedly, mouse liver and primary hepatocytes consumed ketone bodies to support fatty acid biosynthesis via both de novo lipogenesis (DNL) and polyunsaturated fatty acid (PUFA) elongation. While an acetoacetate intermediate was not absolutely required for ketone bodies to source DNL, PUFA elongation required activation of acetoacetate by cytosolic acetoacetyl-coenzyme A synthetase (AACS). Moreover, AACS deficiency diminished free and esterified PUFAs in hepatocytes, while ketogenic insufficiency depleted PUFAs and increased liver triacylglycerols. These findings suggest that hepatic ketogenesis influences PUFA metabolism, representing a molecular mechanism through which ketone bodies could influence systemic physiology and chronic diseases.
Collapse
Affiliation(s)
- Eric D. Queathem
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Zahra Moazzami
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - David B. Stagg
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Kyle Fulghum
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Abdirahman Hayir
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alisha Seay
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jacob R. Gillingham
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - D. André d’Avignon
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xianlin Han
- Department of Medicine-Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter A. Crawford
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
6
|
Muralidharan S, Lee JWJ, Lim YS, Muthiah M, Tan E, Demicioglu D, Shabbir A, Loo WM, Koo CS, Lee YM, Soon G, Wee A, Halisah N, Abbas S, Ji S, Triebl A, Burla B, Koh HWL, Chan YS, Lee MC, Ng HH, Wenk MR, Torta F, Dan YY. Serum lipidomic signatures in patients with varying histological severity of metabolic-dysfunction associated steatotic liver disease. Metabolism 2025; 162:156063. [PMID: 39522592 DOI: 10.1016/j.metabol.2024.156063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/13/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a spectrum of pathologies ranging from simple steatosis to steatohepatitis, fibrosis and cirrhosis. Patients with metabolic associated steatohepatitis (MASH) with fibrosis are at greatest risk of liver and cardiovascular complications. To identify such at-risk MASLD patients, physicians are still reliant on invasive liver biopsies. This study aimed to identify circulating lipidomic signatures to better identify patients with MASH in a multi-ethnic Asian cohort. APPROACH & RESULTS A lipidomic approach was used to quantify a total of 481 serum lipids from 151 Singaporean patients paired with protocolized liver biopsies. Lipidomic signatures for MASLD, at-risk MASH and advanced fibrosis were identified. 210 lipids showed significant differences for varying histological subtypes of MASLD. Majority of these lipids were associated with liver steatosis (198/210). We identified a panel of 13 lipids associated with lobular inflammation, ballooning and significant fibrosis. Of note, dihexosylceramides were novel markers for significant fibrosis. Using the serum lipidome alone, we could stratify patients with MASLD (AUROC 0.863), as well as those with at-risk MASH (AUROC 0.912) and advanced fibrosis (AUROC 0.95). The lipidomic at-risk MASH predictor, using 14 markers, was independently validated (n = 105) with AUROC 0.76. CONCLUSIONS The dynamic shift in serum lipid profile was associated with progressive histological stages of MASLD, providing surrogate markers for distinguishing stages of MASLD as well as identifying novel pathways in the pathogenesis.
Collapse
Affiliation(s)
- Sneha Muralidharan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Jonathan W J Lee
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology & Hepatology, National University Hospital, Singapore; iHealthtech, National University of Singapore, Singapore
| | - Yee Siang Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Eunice Tan
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | | | - Asim Shabbir
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Surgery, National University Hospital, Singapore
| | - Wai Mun Loo
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Chieh Sian Koo
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Yin Mei Lee
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Gwyneth Soon
- Department of Pathology, National University Hospital, Singapore
| | - Aileen Wee
- Department of Pathology, National University Hospital, Singapore; Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nur Halisah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sakinah Abbas
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shanshan Ji
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Alexander Triebl
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Bo Burla
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Hiromi W L Koh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yun Shen Chan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Mei Chin Lee
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Huck Hui Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore; Precision Medicine Translational Research Programme and Department of Biochemistry, National University of Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore; Precision Medicine Translational Research Programme and Department of Biochemistry, National University of Singapore, Singapore; Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Yock Young Dan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology & Hepatology, National University Hospital, Singapore.
| |
Collapse
|
7
|
Shen C, Pan Z, Xie W, Zhao J, Miao D, Zhao L, Liu M, Zhong Y, Zhong C, Gonzalez FJ, Wang W, Gao Y, Liu C. Hepatocyte-specific SLC27A4 deletion ameliorates nonalcoholic fatty liver disease in mice via suppression of phosphatidylcholine-mediated PXR activation. Metabolism 2025; 162:156054. [PMID: 39489412 DOI: 10.1016/j.metabol.2024.156054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/08/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND The protein Solute carrier family 27 member 4 (SLC27A4) is crucial for fatty acid synthesis and β-oxidation, but its role in hepatic steatosis and nonalcoholic fatty liver disease (NAFLD) progression is not fully understood. METHODS Mice with AAV-mediated overexpression of Slc27a4 in liver and hepatocytes-specific deletion of Slc27a4 were fed a standard chow diet, a high-fat diet (HFD), or a methionine and choline-deficient diet (MCD). Serum and liver tissues were collected and analyzed by biochemical assay, histology, lipidomic analysis, RNA-seq analysis, qPCR, western blot and immunofluorescence. RESULTS This study found elevated expression of SLC27A4 in individuals with NAFLD and OAPA-treated MPHs cells, leading to increased lipid accumulation and diet-induced liver steatosis, inflammation, and fibrosis. Conversely, hepatocyte-specific deletion of Slc27a4 improved the development of both NAFLD and NASH. SLC27A4 overexpression resulted in increased hepatic pregnane X receptor (PXR) expression and accumulation of phosphatidylcholine (PC), which activates PXR signaling and inducing SLC27A4 expression. PXR overexpression hinders the protective impact of Slc27a4 deletion on lipid accumulation and inflammation, whereas its deficiency in mice reduces the effect of Slc27a4 overexpression on NAFLD development. CONCLUSION These results indicate that SLC27A4 plays a critical role of lipid accumulation and inflammation, and is implicated in the development of NAFLD progression, rendering it potentially actionable target for NAFLD treatment.
Collapse
Affiliation(s)
- Chuangpeng Shen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405,China; ShenShan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Shanwei 516600,China
| | - Zhisen Pan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wenmin Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510405,China
| | - Jian Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405,China
| | - Deyu Miao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405,China
| | - Ling Zhao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Min Liu
- Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanhua Zhong
- Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chong Zhong
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405,China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510405,China.
| | - Yong Gao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510405,China.
| | - Changhui Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510405,China.
| |
Collapse
|
8
|
Fujimoto K, Kishino H, Hirao J, Maejima T, Mori K, Tsuchiya Y. Male rat-specific fatty change in liver by DS-1971a: Elevation in phospholipids and adenosine as early responses to the fatty change in male rat-derived primary hepatocytes. J Toxicol Sci 2025; 50:125-134. [PMID: 40024756 DOI: 10.2131/jts.50.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
In a 3-month repeated oral dose toxicity study of DS-1971a, a selective inhibitor of the Nav1.7 voltage-gated sodium channel, fatty change of hepatocytes was observed only in male rats at doses of 100 mg/kg and above. However, this change was not observed in female rats even at the highest dose of 1500 mg/kg. Furthermore, fatty change was not observed in mice and monkeys administered the highest dose of 1000 mg/kg for 6 and 9 months, respectively. To further investigate species differences of this fatty change, lipid accumulation was evaluated by staining with the LipidTOX dye in primary cultured hepatocytes derived from male and female rats, male monkeys, and male and female humans. After exposure to DS-1971a for 72 hr, the staining showed an increase in intensity specifically in male rat-derived hepatocytes in a concentration-dependent manner. Metabolomic analysis using rat-derived primary cultured hepatocytes exposed to DS-1971a for 24 and 72 hr revealed that phospholipids, not neutral lipids like triacylglycerols, and adenosine were elevated in the male-derived hepatocytes. These results suggest that the elevation of phospholipids and adenosine in the hepatocytes may contribute to the specific fatty change observed in male rats.
Collapse
Affiliation(s)
- Kazunori Fujimoto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
- Tissue and Cell Research Center Munich, Daiichi Sankyo Europe GmbH, Germany
| | | | - Jun Hirao
- Regulatory Affairs Strategy Department, Daiichi Sankyo Co., Ltd
| | | | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | | |
Collapse
|
9
|
Flam E, Haas JT, Staels B. Liver metabolism in human MASLD: A review of recent advancements using human tissue metabolomics. Atherosclerosis 2025; 400:119054. [PMID: 39586140 DOI: 10.1016/j.atherosclerosis.2024.119054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024]
Abstract
Global incidence of Metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) is on the rise while treatments remain elusive. MASLD is a disease of dysregulated systemic and hepatic metabolism. Current understanding of disease pathophysiology as it relates to metabolome changes largely comes from studies on animal models and human plasma. However, human tissue data are crucial for transitioning from mechanisms to clinical therapies. The close relationship between MASLD and comorbidities like obesity, type 2 diabetes and dyslipidemia make it difficult to determine the contribution from liver disease itself. Here, we review recent metabolomics studies in liver tissue from human MASLD patients, which have predominately focused on lipid metabolism, but also include bile acid, tricarboxylic acid (TCA) cycle, and branched chain amino acid (BCAA) metabolism. Several clinical trials are underway to target various of these lipid-related pathways in MASLD. Although only the β-selective thyroid hormone receptor agonist resmetirom has so far been approved for use, many metabolism-targeting pharmaceuticals show promising results for halting disease progression, if not promoting outright reversal. Ultimately, the scarcity of human tissue data and the variability of confounding factors, like obesity, within and between cohorts are impediments to the pathophysiological understanding required for efficient development of metabolic treatments.
Collapse
Affiliation(s)
- Emily Flam
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
10
|
Queathem ED, Stagg D, Nelson A, Chaves AB, Crown SB, Fulghum K, D Avignon DA, Ryder JR, Bolan PJ, Hayir A, Gillingham JR, Jannatpour S, Rome FI, Williams AS, Muoio DM, Ikramuddin S, Hughey CC, Puchalska P, Crawford PA. Ketogenesis protects against MASLD-MASH progression through mechanisms that extend beyond overall fat oxidation rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618895. [PMID: 39464122 PMCID: PMC11507910 DOI: 10.1101/2024.10.17.618895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The progression of metabolic-dysfunction-associated steatotic liver disease (MASLD) to metabolic-dysfunction-associated steatohepatitis (MASH) involves complex alterations in both liver-autonomous and systemic metabolism that influence the liver's balance of fat accretion and disposal. Here, we quantify the relative contribution of hepatic oxidative pathways to liver injury in MASLD-MASH. Using NMR spectroscopy, UHPLC-MS, and GC-MS, we performed stable-isotope tracing and formal flux modeling to quantify hepatic oxidative fluxes in humans across the spectrum of MASLD-MASH, and in mouse models of impaired ketogenesis. We found in humans with MASH, that liver injury correlated positively with ketogenesis and total fat oxidation, but not with turnover of the tricarboxylic acid cycle. The use of loss-of-function mouse models demonstrated that disruption of mitochondrial HMG-CoA synthase (HMGCS2), the rate-limiting step of ketogenesis, impairs overall hepatic fat oxidation and induces a MASLD-MASH-like phenotype. Disruption of mitochondrial β-hydroxybutyrate dehydrogenase (BDH1), the terminal step of ketogenesis, also impaired fat oxidation, but surprisingly did not exacerbate steatotic liver injury. Taken together, these findings suggest that quantifiable variations in overall hepatic fat oxidation may not be a primary determinant of MASLD-to-MASH progression, but rather, that maintenance of hepatic ketogenesis could serve a protective role through additional mechanisms that extend beyond quantified overall rates of fat oxidation.
Collapse
|
11
|
Gautam J, Aggarwal H, Kumari D, Gupta SK, Kumar Y, Dikshit M. A methionine-choline-deficient diet induces nonalcoholic steatohepatitis and alters the lipidome, metabolome, and gut microbiome profile in the C57BL/6J mouse. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159545. [PMID: 39089643 DOI: 10.1016/j.bbalip.2024.159545] [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: 02/12/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The methionine-choline-deficient (MCD) diet-induced non-alcoholic steatohepatitis (NASH) in mice is a well-established model. Our study aims to elucidate the factors influencing liver pathology in the MCD mouse model by examining physiological, biochemical, and molecular changes using histology, molecular techniques, and OMICS approaches (lipidomics, metabolomics, and metagenomics). Male C57BL/6J mice were fed a standard chow diet, a methionine-choline-sufficient (MCS) diet, or an MCD diet for 10 weeks. The MCD diet resulted in reduced body weight and fat mass, along with decreased plasma triglyceride, cholesterol, glucose, and insulin levels. However, it notably induced steatosis, inflammation, and alterations in gene expression associated with lipogenesis, inflammation, fibrosis, and the synthesis of apolipoproteins, sphingolipids, ceramides, and carboxylesterases. Lipid analysis revealed significant changes in plasma and tissues: most ceramide non-hydroxy-sphingosine lipids significantly decreased in the liver and plasma but increased in the adipose tissue of MCD diet-fed animals. Oxidized glycerophospholipids mostly increased in the liver but decreased in the adipose tissue of the MCD diet-fed group. The gut microbiome of the MCD diet-fed group showed an increase in Firmicutes and a decrease in Bacteroidetes and Actinobacteria. Metabolomic profiling demonstrated that the MCD diet significantly altered amino acid biosynthesis, metabolism, and nucleic acid metabolism pathways in plasma, liver, fecal, and cecal samples. LC-MS data indicated higher total plasma bile acid intensity and reduced fecal glycohyodeoxycholic acid intensity in the MCD diet group. This study demonstrates that although the MCD diet induces hepatic steatosis, the mechanisms underlying NASH in this model differ from those in human NASH pathology.
Collapse
Affiliation(s)
- Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
12
|
Tao Y, Chen Y, Ren J, Jiang S, Zhang S, Xu H, Li Y. Lipidomics and transcriptomics analysis revealed the role of the spleen of Nile tilapia (Oreochromis niloticus) in lipid metabolism. AQUACULTURE 2024; 592:741173. [DOI: 10.1016/j.aquaculture.2024.741173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
López-Yerena A, Muñoz-García N, de Santisteban Villaplana V, Padro T, Badimon L. Effect of Moderate Beer Intake on the Lipid Composition of Human Red Blood Cell Membranes. Nutrients 2024; 16:3541. [PMID: 39458535 PMCID: PMC11510343 DOI: 10.3390/nu16203541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Growing evidence suggests that erythrocyte membrane lipids are subject to changes during their lifespan. Factors such as the type of dietary intake and its composition contribute to the changes in red blood cell (RBC) membranes. Due to the high antioxidant content of beer, we aimed to investigate the effect of moderate beer consumption on the lipid composition of RBCs membranes from healthy overweight individuals. Methods: We conducted a four-weeks, prospective two-arm longitudinal crossed-over study, where participants (n = 36) were randomly assigned to alcohol-free beer group or traditional beer group. The lipids of RBCs membranes were assessed at the beginning and the end of the intervention by thin-layer chromatography. Results: Four-weeks of alcohol-free beer promoted changes in fatty acids (FA), free cholesterol (FC), phosphatidylethanolamine (PE) and phosphatidylcholine (PC) (p < 0.05). Meanwhile, traditional beer intake led to changes in FA, FC, phospholipids (PL), PE and PC (p < 0.05). The observed alterations in membrane lipids were found to be independent of sex and BMI as influencing factors. Conclusions: The lipid composition of erythrocyte membranes is distinctly but mildly influenced by the consumption of both non-alcoholic and conventional beer, with no effects on RBC membrane fluidity.
Collapse
Affiliation(s)
- Anallely López-Yerena
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
| | - Natalia Muñoz-García
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
| | - Victoria de Santisteban Villaplana
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- School of Pharmacy and Food Sciences, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Teresa Padro
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Lina Badimon
- Institut Recerca Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; (A.L.-Y.); (N.M.-G.); (V.d.S.V.); (T.P.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| |
Collapse
|
14
|
Fan X, Hu X, Cong P, Wang X, Song Y, Liu Y, Wang X, Meng N, Xue C, Xu J. Combined UPLC-QqQ-MS/MS and AP-MALDI Mass Spectrometry Imaging Method for Phospholipidomics in Obese Mouse Kidneys: Alleviation by Feeding Sea Cucumber Phospholipids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16312-16322. [PMID: 38985073 DOI: 10.1021/acs.jafc.4c02692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Sea cucumber phospholipids have ameliorative effects on various diseases related to lipid metabolism. However, it is unclear whether it can ameliorate obesity-associated glomerulopathy (ORG) induced by a high-fat diet (HFD). The present study applied UPLC-QqQ-MS/MS and atmospheric pressure matrix-assisted laser desorption ionization mass spectrometry imaging (AP-MALDI MSI) to investigate the effects of sea cucumber phospholipids, including plasmalogen PlsEtn and plasmanylcholine PakCho, on phospholipid profiles in the HFD-induced ORG mouse kidney. Quantitative analysis of 135 phospholipids revealed that PlsEtn and PakCho significantly modulated phospholipid levels. Notably, PlsEtn modulated kidney overall phospholipids better than PakCho. Imaging the "space-content" of 9 phospholipids indicated that HFD significantly increased phospholipid content within the renal cortex. Furthermore, PlsEtn and PakCho significantly decreased the expression of transport-related proteins CD36, while elevating the expression of fatty acid β-oxidation-related protein PPAR-α in the renal cortex. In conclusion, sea cucumber phospholipids reduced renal lipid accumulation, ameliorated renal damage, effectively regulated the content and distribution of renal phospholipids, and improved phospholipid homeostasis, exerting an anti-OGR effect.
Collapse
Affiliation(s)
- Xiaowei Fan
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Xinxin Hu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Peixu Cong
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Xincen Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
- Institute of Nutrition and Health, Qingdao University, Qingdao, Shandong 266073, China
| | - Yu Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Yanjun Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Xiaoxu Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Nan Meng
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
- Qingdao Marine Science and Technology Center, Qingdao, Shandong 266235, China
| | - Jie Xu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266404, China
| |
Collapse
|
15
|
Miller JM, Tragesser-Tiña ME, Turk SM, Rubenstein EM. Loss of transcriptional regulator of phospholipid biosynthesis alters post-translational modification of Sec61 translocon beta subunit Sbh1 in Saccharomyces cerevisiae. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001260. [PMID: 39071171 PMCID: PMC11282434 DOI: 10.17912/micropub.biology.001260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
We recently discovered that disrupting phospholipid biosynthesis by eliminating the Ino2/4 transcriptional regulator impairs endoplasmic reticulum (ER)-associated degradation (ERAD) in Saccharomyces cerevisiae , but the mechanism is unclear. Phosphatidylcholine deficiency has been reported to accelerate degradation of Sec61 translocon beta subunit Sbh1 and ERAD cofactor Cue1. Here, we found that, unlike targeted phosphatidylcholine depletion, INO4 deletion does not destabilize Sbh1 or Cue1. However, we observed altered electrophoretic mobility of Sbh1 in ino4 Δ yeast, consistent with phospholipid-responsive post-translational modification. A better understanding of the molecular consequences of disrupted lipid homeostasis could lead to enhanced treatments for conditions associated with perturbed lipid biosynthesis.
Collapse
Affiliation(s)
| | - Mary E. Tragesser-Tiña
- Department of Biology, Ball State University
- Diabetes, Obesity, and Complications Therapeutic Area, Eli Lilly and Company
| | - Samantha M. Turk
- Department of Biology, Ball State University
- Graduate School of Biomedical Sciences and Department of Developmental Neurobiology, St. Jude Graduate School of Biomedical Science
| | | |
Collapse
|
16
|
Queathem ED, Moazzami Z, Stagg DB, Nelson AB, Fulghum K, Hayir A, Seay A, Gillingham JR, d'Avignon DA, Han X, Ruan HB, Crawford PA, Puchalska P. Ketogenesis supports hepatic polyunsaturated fatty acid homeostasis via fatty acid elongation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602593. [PMID: 39026753 PMCID: PMC11257565 DOI: 10.1101/2024.07.09.602593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Therapeutic interventions targeting hepatic lipid metabolism in metabolic dysfunction-associated steatotic liver disease (MASLD) and steatohepatitis (MASH) remain elusive. Using mass spectrometry-based stable isotope tracing and shotgun lipidomics, we established a novel link between ketogenesis and MASLD pathophysiology. Our findings show that mouse liver and primary hepatocytes consume ketone bodies to support fatty acid (FA) biosynthesis via both de novo lipogenesis (DNL) and FA elongation. Analysis of 13 C-labeled FAs in hepatocytes lacking mitochondrial D-β-hydroxybutyrate dehydrogenase (BDH1) revealed a partial reliance on mitochondrial conversion of D-βOHB to acetoacetate (AcAc) for cytoplasmic DNL contribution, whereas FA elongation from ketone bodies was fully dependent on cytosolic acetoacetyl-CoA synthetase (AACS). Ketone bodies were essential for polyunsaturated FA (PUFA) homeostasis in hepatocytes, as loss of AACS diminished both free and esterified PUFAs. Ketogenic insufficiency depleted liver PUFAs and increased triacylglycerols, mimicking human MASLD, suggesting that ketogenesis supports PUFA homeostasis, and may mitigate MASLD-MASH progression in humans.
Collapse
|
17
|
Chowdhury RR, Grosso MF, Gadara DC, Spáčil Z, Vidová V, Sovadinová I, Babica P. Cyanotoxin cylindrospermopsin disrupts lipid homeostasis and metabolism in a 3D in vitro model of the human liver. Chem Biol Interact 2024; 397:111046. [PMID: 38735451 DOI: 10.1016/j.cbi.2024.111046] [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: 08/20/2023] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Cylindrospermopsin, a potent hepatotoxin produced by harmful cyanobacterial blooms, poses environmental and human health concerns. We used a 3D human liver in vitro model based on spheroids of HepG2 cells, in combination with molecular and biochemical assays, automated imaging, targeted LC-MS-based proteomics, and lipidomics, to explore cylindrospermopsin effects on lipid metabolism and the processes implicated in hepatic steatosis. Cylindrospermopsin (1 μM, 48 h) did not significantly affect cell viability but partially reduced albumin secretion. However, it increased neutral lipid accumulation in HepG2 spheroids while decreasing phospholipid levels. Simultaneously, cylindrospermopsin upregulated genes for lipogenesis regulation (SREBF1) and triacylglycerol synthesis (DGAT1/2) and downregulated genes for fatty acid synthesis (ACLY, ACCA, FASN, SCD1). Fatty acid uptake, oxidation, and lipid efflux genes were not significantly affected. Targeted proteomics revealed increased levels of perilipin 2 (adipophilin), a major hepatocyte lipid droplet-associated protein. Lipid profiling quantified 246 lipid species in the spheroids, with 28 significantly enriched and 15 downregulated by cylindrospermopsin. Upregulated species included neutral lipids, sphingolipids (e.g., ceramides and dihexosylceramides), and some glycerophospholipids (phosphatidylethanolamines, phosphatidylserines), while phosphatidylcholines and phosphatidylinositols were mostly reduced. It suggests that cylindrospermopsin exposures might contribute to developing and progressing towards hepatic steatosis or metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Riju Roy Chowdhury
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Marina Felipe Grosso
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | | | - Zdeněk Spáčil
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Veronika Vidová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Iva Sovadinová
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kotlářská 2, Brno, Czech Republic.
| |
Collapse
|
18
|
Ma Z, He Y, Li Y, Wang Q, Fang M, Yang Q, Gong Z, Xu L. Effects of Deoxynivalenol and Its Acetylated Derivatives on Lipid Metabolism in Human Normal Hepatocytes. Toxins (Basel) 2024; 16:294. [PMID: 39057934 PMCID: PMC11281666 DOI: 10.3390/toxins16070294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Deoxynivalenol (DON), 3-acetyldeoxynivalenol (3-ADON) and 15-acetyldeoxynivalenol (15-ADON) belong to type B trichothecenes that are widely detected in agricultural products as one of the most common classes of mycotoxins. In the present study, we aimed to characterize the alteration of lipid metabolism in normal human hepatocytes by poisoning with DON and its acetylated derivatives. After verifying the hepatotoxicity of the three toxins, DON, 15-ADON, and 3-ADON, the mRNA expression was determined by transcriptomics, and the results showed that DON and 15-ADON had a significant regulatory effect on the transcriptome, in which glycerophospholipid metabolism pathway and phospholipase D signaling pathways have not been reported in studies of DON and its acetylated derivatives. For further validation, we explored lipid metabolism in depth and found that PC (15:0/16:0), PC (16:1/18:3), PC (18:1/22:6), PC (16:0/16:0), PC (16:0/16:1), PC (16:1/18:1), PC (14:0/18:2), PE (14:0/16:0) and PE (18:1/18:3) were downregulated for all nine lipids. Combined with the transcriptome results, we found that hepatic steatosis induced by the three toxins, DON, 15-ADON and 3-ADON, was associated with altered expression of genes related to lipid oxidation, lipogenesis and lipolysis, and their effects on lipid metabolism in L-02 cells were mainly realized through the PC-PE cycle.
Collapse
Affiliation(s)
- Zhaoqing Ma
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuyun He
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuzhi Li
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
- Key Laboratory of Detection Technology of Focus Chemical Hazards in Animal-Derived Food for State Market Regulation, Wuhan 430075, China
| | - Qiao Wang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Min Fang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Qing Yang
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Zhiyong Gong
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| | - Lin Xu
- College of Food Scienceand Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan 430023, China
| |
Collapse
|
19
|
Yang H, Suh DH, Jung ES, Lee Y, Liu KH, Do IG, Lee CH, Park CY. Ezetimibe, Niemann-Pick C1 like 1 inhibitor, modulates hepatic phospholipid metabolism to alleviate fat accumulation. Front Pharmacol 2024; 15:1406493. [PMID: 38953111 PMCID: PMC11215075 DOI: 10.3389/fphar.2024.1406493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 07/03/2024] Open
Abstract
Background Ezetimibe, which lowers cholesterol by blocking the intestinal cholesterol transporter Niemann-Pick C1 like 1, is reported to reduce hepatic steatosis in humans and animals. Here, we demonstrate the changes in hepatic metabolites and lipids and explain the underlying mechanism of ezetimibe in hepatic steatosis. Methods We fed Otsuka Long-Evans Tokushima Fatty (OLETF) rats a high-fat diet (60 kcal % fat) with or vehicle (control) or ezetimibe (10 mg kg-1) via stomach gavage for 12 weeks and performed comprehensive metabolomic and lipidomic profiling of liver tissue. We used rat liver tissues, HepG2 hepatoma cell lines, and siRNA to explore the underlying mechanism. Results In OLETF rats on a high-fat diet, ezetimibe showed improvements in metabolic parameters and reduction in hepatic fat accumulation. The comprehensive metabolomic and lipidomic profiling revealed significant changes in phospholipids, particularly phosphatidylcholines (PC), and alterations in the fatty acyl-chain composition in hepatic PCs. Further analyses involving gene expression and triglyceride assessments in rat liver tissues, HepG2 hepatoma cell lines, and siRNA experiments unveiled that ezetimibe's mechanism involves the upregulation of key phospholipid biosynthesis genes, CTP:phosphocholine cytidylyltransferase alpha and phosphatidylethanolamine N-methyl-transferase, and the phospholipid remodeling gene lysophosphatidylcholine acyltransferase 3. Conclusion This study demonstrate that ezetimibe improves metabolic parameters and reduces hepatic fat accumulation by influencing the composition and levels of phospholipids, specifically phosphatidylcholines, and by upregulating genes related to phospholipid biosynthesis and remodeling. These findings provide valuable insights into the molecular pathways through which ezetimibe mitigates hepatic fat accumulation, emphasizing the role of phospholipid metabolism.
Collapse
Affiliation(s)
- Hyekyung Yang
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Ho Suh
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Eun Sung Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Yoonjin Lee
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kwang-Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - In-Gu Do
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Cheol-Young Park
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Qi Z, LE S, Cheng R, DU X, Zhao C, Zhang Z, Zhang X, Feng L, Schumann M, Mao L, Cheng S. Responses of the Serum Lipid Profile to Exercise and Diet Interventions in Nonalcoholic Fatty Liver Disease. Med Sci Sports Exerc 2024; 56:1036-1045. [PMID: 38247038 DOI: 10.1249/mss.0000000000003388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
BACKGROUND This study aimed to assess the response patterns of circulating lipids to exercise and diet interventions in nonalcoholic fatty liver disease (NAFLD). METHODS The 8.6-month four-arm randomized controlled study comprised 115 NAFLD patients with prediabetes who were assigned to aerobic exercise (AEx; n = 29), low-carbohydrate diet (Diet; n = 28), AEx plus low-carbohydrate diet (AED; n = 29), and nonintervention (NI, n = 29) groups. Hepatic fat content (HFC) was quantified by proton magnetic resonance spectroscopy. Serum lipidomic analytes were measured using liquid chromatography-mass spectrometry. RESULTS After intervention, the total level of phosphatidylcholine (PC) increased significantly in the AEx group ( P = 0.043), whereas phosphatidylethanolamine (PE) and triacylglycerol decreased significantly in the AED group ( P = 0.046 and P = 0.036, respectively), and phosphatidylserine decreased in the NI group ( P = 0.002). Changes of 21 lipid metabolites were significantly associated with changes of HFC, among which half belonged to PC. Most of the molecules related to insulin sensitivity belonged to sphingomyelin (40 of 79). Controlling for the change of visceral fat, the significant associations between lipid metabolites and HFC remained. In addition, baseline serum lipids could predict the response of HFC to exercise and/or diet interventions (PE15:0/18:0 for AED, area under the curve (AUC) = 0.97; PE22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0 for AEx, AUC = 0.90; and PC14:1(9Z)/19:1(9Z) for Diet, AUC = 0.92). CONCLUSIONS Changes of lipidome after exercise and/or diet interventions were associated with HFC reductions, which are independent of visceral fat reduction, particularly in metabolites belonging to PC. Importantly, baseline PE could predict the HFC response to exercise, and PC predicted the response to diet. These results indicate that a circulating metabolomics panel can be used to facilitate clinical implementation of lifestyle interventions for NAFLD management.
Collapse
Affiliation(s)
- Zhen Qi
- Physical Education Department, Shanghai Jiao Tong University, Shanghai, CHINA
| | | | - Runtan Cheng
- Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, CHINA
| | - Xiaming DU
- Department of Orthopedic, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, CHINA
| | - Can Zhao
- School of Athletic Performance, Shanghai University of Sport, Shanghai, CHINA
| | - Zhengyun Zhang
- Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA
| | - Xiaobo Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital of Wenzhou Medical University, Wenzhou, CHINA
| | - Lei Feng
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, CHINA
| | | | - Lijuan Mao
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai 200438, CHINA
| | | |
Collapse
|
21
|
Song Q, Zhao Z, Liu H, Zhang J, Wang Z, Zhang Y, Ma G, Ge S. Pseudotargeted lipidomics analysis of scoparone on glycerophospholipid metabolism in non-alcoholic steatohepatitis mice by LC-MRM-MS. PeerJ 2024; 12:e17380. [PMID: 38799063 PMCID: PMC11122033 DOI: 10.7717/peerj.17380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/21/2024] [Indexed: 05/29/2024] Open
Abstract
As the inflammatory subtype of nonalcoholic fatty liver disease (NAFLD), the progression of nonalcoholic steatohepatitis (NASH) is associated with disorders of glycerophospholipid metabolism. Scoparone is the major bioactive component in Artemisia capillaris which has been widely used to treat NASH in traditional Chinese medicine. However, the underlying mechanisms of scoparone against NASH are not yet fully understood, which hinders the development of effective therapeutic agents for NASH. Given the crucial role of glycerophospholipid metabolism in NASH progression, this study aimed to characterize the differential expression of glycerophospholipids that is responsible for scoparone's pharmacological effects and assess its efficacy against NASH. Liquid chromatography-multiple reaction monitoring-mass spectrometry (LC-MRM-MS) was performed to get the concentrations of glycerophospholipids, clarify mechanisms of disease, and highlight insights into drug discovery. Additionally, pathologic findings also presented consistent changes in high-fat diet-induced NASH model, and after scoparone treatment, both the levels of glycerophospholipids and histopathology were similar to normal levels, indicating a beneficial effect during the observation time. Altogether, these results refined the insights on the mechanisms of scoparone against NASH and suggested a route to relieve NASH with glycerophospholipid metabolism. In addition, the current work demonstrated that a pseudotargeted lipidomic platform provided a novel insight into the potential mechanism of scoparone action.
Collapse
Affiliation(s)
- Qi Song
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ziyi Zhao
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Hu Liu
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Jinling Zhang
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Zhiqiang Wang
- Hebei Key Laboratory of Public Health Safety, School of Public HealthPublic Health, Hebei University, Baoding, Hebei, China
| | - Yunqi Zhang
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Guowei Ma
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Shaoqin Ge
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
- College of Basic Medical Science, Hebei University of Technology, Baoding, Hebei, China
| |
Collapse
|
22
|
Gu TJ, Liu PK, Wang YW, Flowers MT, Xu S, Liu Y, Davis DB, Li L. Diazobutanone-assisted isobaric labelling of phospholipids and sulfated glycolipids enables multiplexed quantitative lipidomics using tandem mass spectrometry. Nat Chem 2024; 16:762-770. [PMID: 38365942 DOI: 10.1038/s41557-023-01436-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 12/21/2023] [Indexed: 02/18/2024]
Abstract
Mass spectrometry-based quantitative lipidomics is an emerging field aiming to uncover the intricate relationships between lipidomes and disease development. However, quantifying lipidomes comprehensively in a high-throughput manner remains challenging owing to the diverse lipid structures. Here we propose a diazobutanone-assisted isobaric labelling strategy as a rapid and robust platform for multiplexed quantitative lipidomics across a broad range of lipid classes, including various phospholipids and glycolipids. The diazobutanone reagent is designed to conjugate with phosphodiester or sulfate groups, while accommodating various functional groups on different lipid classes, enabling subsequent isobaric labelling for high-throughput multiplexed quantitation. Our method demonstrates excellent performance in terms of labelling efficiency, detection sensitivity, quantitative accuracy and broad applicability to various biological samples. Finally, we performed a six-plex quantification analysis of lipid extracts from lean and obese mouse livers. In total, we identified and quantified 246 phospholipids in a high-throughput manner, revealing lipidomic changes that may be associated with obesity in mice.
Collapse
Affiliation(s)
- Ting-Jia Gu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Peng-Kai Liu
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yen-Wen Wang
- Department of Biostatics, Yale University, New Haven, CT, USA
| | - Matthew T Flowers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Shuling Xu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuan Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Dawn B Davis
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Chi L, YifeiYang, Bian X, Gao B, Tu P, Ru H, Lu K. Chronic sucralose consumption inhibits farnesoid X receptor signaling and perturbs lipid and cholesterol homeostasis in the mouse livers, potentially by altering gut microbiota functions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:169603. [PMID: 38272087 DOI: 10.1016/j.scitotenv.2023.169603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024]
Abstract
Sucralose has raised concerns regarding its safety and recent studies have demonstrated that sucralose consumption can disrupt the normal gut microbiome and alter metabolic profiles in mice. However, the extent to which this perturbation affects the functional interaction between the microbiota and the host, as well as its potential impact on host health, remains largely unexplored. Here, we aimed to investigate whether chronic sucralose consumption, at levels within the Acceptable Daily Intake (ADI), could disturb key gut microbial functions and lead to adverse health effects in mice. Following six-month sucralose consumption, several bacterial genera associated with bile acid metabolism were decreased, including Lactobacillus and Ruminococcus. Consequently, the richness of secondary bile acid biosynthetic pathway and bacterial bile salt hydrolase gene were decreased in the sucralose-treated gut microbiome. Compared to controls, sucralose-consuming mice exhibited significantly lower ratios of free bile acids and taurine-conjugated bile acids in their livers. Additionally, several farnesoid X receptor (FXR) agonists were decreased in sucralose-treated mice. This reduction in hepatic FXR activation was associated with altered expression of down-stream genes, in the liver. Moreover, the expression of key lipogenic genes was up-regulated in the livers of sucralose-treated mice. Changes in hepatic lipid profiles were also observed, characterized by lower ceramide levels, a decreased PC/PE ratio, and a mildly increase in lipid accumulation. Additionally, sucralose-consumed mice exhibited higher hepatic cholesterol level compared to control mice, with up-regulation of cholesterol efflux genes and down-regulation of genes associated with reverse cholesterol transport. In conclusion, chronic sucralose consumption disrupts FXR signaling activation and perturbs hepatic lipid and cholesterol homeostasis, potentially by diminishing the bile acid metabolic capacity of the gut microbiome. These findings shed light on the complex interplay between sucralose, the gut microbiota, and host metabolism, raising important questions about the safety of its long-term consumption.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC 27599, United States
| | - YifeiYang
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Xiaoming Bian
- Department of Environmental Health Sciences, University of Georgia, Athens, GA, 30602, United States of America
| | - Bei Gao
- Department of Environmental Health Sciences, University of Georgia, Athens, GA, 30602, United States of America
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Hongyu Ru
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC 27599, United States; Institute for Environmental Health Solutions, University of North Carolina at Chapel Hill, NC 27599, United States.
| |
Collapse
|
24
|
Rodrigues SG, van der Merwe S, Krag A, Wiest R. Gut-liver axis: Pathophysiological concepts and medical perspective in chronic liver diseases. Semin Immunol 2024; 71:101859. [PMID: 38219459 DOI: 10.1016/j.smim.2023.101859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/11/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024]
Affiliation(s)
- Susana G Rodrigues
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Schalk van der Merwe
- Department of Gastroenterology and Hepatology, University hospital Gasthuisberg, University of Leuven, Belgium
| | - Aleksander Krag
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark, University of Southern Denmark, Odense, Denmark
| | - Reiner Wiest
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| |
Collapse
|
25
|
Sun C, Lan F, Zhou Q, Guo X, Jin J, Wen C, Guo Y, Hou Z, Zheng J, Wu G, Li G, Yan Y, Li J, Ma Q, Yang N. Mechanisms of hepatic steatosis in chickens: integrated analysis of the host genome, molecular phenomics and gut microbiome. Gigascience 2024; 13:giae023. [PMID: 38837944 PMCID: PMC11152177 DOI: 10.1093/gigascience/giae023] [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: 05/14/2023] [Revised: 01/14/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024] Open
Abstract
Hepatic steatosis is the initial manifestation of abnormal liver functions and often leads to liver diseases such as nonalcoholic fatty liver disease in humans and fatty liver syndrome in animals. In this study, we conducted a comprehensive analysis of a large chicken population consisting of 705 adult hens by combining host genome resequencing; liver transcriptome, proteome, and metabolome analysis; and microbial 16S ribosomal RNA gene sequencing of each gut segment. The results showed the heritability (h2 = 0.25) and duodenal microbiability (m2 = 0.26) of hepatic steatosis were relatively high, indicating a large effect of host genetics and duodenal microbiota on chicken hepatic steatosis. Individuals with hepatic steatosis had low microbiota diversity and a decreased genetic potential to process triglyceride output from hepatocytes, fatty acid β-oxidation activity, and resistance to fatty acid peroxidation. Furthermore, we revealed a molecular network linking host genomic variants (GGA6: 5.59-5.69 Mb), hepatic gene/protein expression (PEMT, phosphatidyl-ethanolamine N-methyltransferase), metabolite abundances (folate, S-adenosylmethionine, homocysteine, phosphatidyl-ethanolamine, and phosphatidylcholine), and duodenal microbes (genus Lactobacillus) to hepatic steatosis, which could provide new insights into the regulatory mechanism of fatty liver development.
Collapse
Affiliation(s)
- Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Xiaoli Guo
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Jiaming Jin
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Yanxin Guo
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Zhuocheng Hou
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Jiangxia Zheng
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Guiqin Wu
- Beijing Engineering Research Centre of Layer,
Beijing 101206, China
| | - Guangqi Li
- Beijing Engineering Research Centre of Layer,
Beijing 101206, China
| | - Yiyuan Yan
- Beijing Engineering Research Centre of Layer,
Beijing 101206, China
| | - Junying Li
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Qiugang Ma
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding, Department of Animal
Genetics and Breeding, College of Animal Science and Technology, China Agricultural
University, Beijing 100193, China
| |
Collapse
|
26
|
Wang X, Wu L, Tao J, Ye H, Wang J, Gao R, Liu W. A lipidomic approach to bisphenol F-induced non-alcoholic fatty liver disease-like changes: altered lipid components in a murine model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:112644-112659. [PMID: 37837594 DOI: 10.1007/s11356-023-30306-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Bisphenol A (BPA), a typical environmental endocrine disruptor, is an "obesogen" that can induce lipid accumulation in the liver. Highly similar in structure to BPA, bisphenol F (BPF) is becoming the dominant BPA substitute on the market, which attracts more and more attention due to its potential adverse effects. Recently, BPF exposure is found to cause non-alcoholic fatty liver disease (NAFLD)-like changes; however, the underlying toxic effects remain poorly understood. Therefore, in the current study, we focused on BPF-mediated lipid homeostasis, especially the alterations of lipid components and metabolism. In human serum, the BPF levels in healthy controls and NAFLD patients were assessed by ELISA, and BPF-induced disturbance of lipid metabolism was evaluated in mouse model via non-targeted lipomic methods with ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry. It suggested that BPF exposure was positively correlated with NAFLD severity and triglyceride level in patients. Based on the relationships, lipid metabolites were assessed in mouse livers between control and BPF-treated group, and it revealed that twenty-six lipid metabolites (including phospholipids, sphingolipids, and glycerides) were significantly changed in mouse livers. Phosphatidylcholine, phosphatidylethanolamine, and diglyceryl ester levels were lower than those in the control mice; hexose ceramide content in sphingolipids markedly increased in BPF-treated mouse livers. Noteworthily, the glycerophospholipid metabolic pathway was found to be the most pronounced in BPF-induced disturbance of lipid metabolism. Therefore, the current study, for the first time, is deciphering the BPF-induced lipid metabolic disturbance, which may provide novel intervention strategies for BPF-induced NAFLD-like changes.
Collapse
Affiliation(s)
- Xinjing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Linlin Wu
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Jingxian Tao
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Heyong Ye
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Jun Wang
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Rong Gao
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
- Department of Hygienic Analysis and Detection, Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenwei Liu
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
27
|
Chen H, Deng Y, Wang Q, Chen W, Liu Z, Tan H, Chen D. Large polystyrene microplastics results in hepatic lipotoxicity in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 333:122015. [PMID: 37343913 DOI: 10.1016/j.envpol.2023.122015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/15/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023]
Abstract
Like small microplastics (MPs), recent studies reveal that large MPs could cause health risks in mice, even if they are not enriched in tissues. However, potential hepatoxicity following large MPs exposure and the underlying mechanisms have not been thoroughly investigated. In the present study, we explored the disruption of hepatic lipid metabolism and potential underlying toxic mechanisms in mice caused by long-term exposure to large polystyrene MPs (40-100 μm) based on a multi-omic approach. After 21 weeks of feeding foods containing MPs (50 and 500 mg/kg food), lipidomic revealed that environmentally relevant and higher doses MP exposures resulted in significant changes in a total of 20 lipid classes. Ceramide (Cer) and dihydroceramide (dhCer) were significantly reduced, while cholesteryl ester (CE), lysoalkylphosphatidylcholine (LPCO), lysophosphatidylethanolamine (LPE) and total glyceride (TG) were all elevated by MPs. The transcriptomic and other physiological data suggested that the potential toxic mechanisms may be related to disorders of fatty acid and cholesterol synthesis and metabolism disorders, and transporting of TG. Our findings demonstrate the hepatic lipotoxicity following exposure to environmentally relevant and higher doses of large MPs, calling for future research and management of the environmental risks of MPs with relatively large particle sizes.
Collapse
Affiliation(s)
- Hexia Chen
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| | - Yongfeng Deng
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China.
| | - Qing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhiteng Liu
- Shenzhen Colleage of International Education, Shenzhen 518043, China
| | - Hongli Tan
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Da Chen
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
| |
Collapse
|
28
|
Araújo COD, Pedroso AP, Boldarine VT, Fernandes AMAP, Perez JJM, Montenegro RM, Montenegro APDR, de Carvalho AB, Fernandes VO, Oyama LM, Carvalho PO, Maia CSC, Bueno AA, Ribeiro EB, Telles MM. Plasma signatures of Congenital Generalized Lipodystrophy patients identified by untargeted lipidomic profiling are not changed after a fat-containing breakfast meal. Prostaglandins Leukot Essent Fatty Acids 2023; 196:102584. [PMID: 37573715 DOI: 10.1016/j.plefa.2023.102584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND The incapacity to store lipids in adipose tissue in Congenital Generalized Lipodystrophy (CGL) causes hypoleptinemia, increased appetite, ectopic fat deposition and lipotoxicity. CGL patients experience shortened life expectancy. The plasma lipidomic profile has not been characterized fully in CGL, nor has the extent of dietary intake in its modulation. The present work investigated the plasma lipidomic profile of CGL patients in comparison to eutrophic individuals at the fasted state and after a breakfast meal. METHOD Blood samples from 11 CGL patients and 10 eutrophic controls were collected after 12 h fasting (T0) and 90 min after an ad libitum fat-containing breakfast (T90). The lipidomic profile of extracted plasma lipids was characterized by non-target liquid chromatography mass spectrometry. RESULTS Important differences between groups were observed at T0 and at T90. Several molecular species of fatty acyls, glycerolipids, sphingolipids and glycerophospholipids were altered in CGL. All the detected fatty acyl molecular species, several diacylglycerols and one triacylglycerol species were upregulated in CGL. Among sphingolipids, one sphingomyelin and one glycosphingolipid species showed downregulation in CGL. Alterations in the glycerophospholipids glycerophosphoethanolamines, glycerophosphoserines and cardiolipins were more complex. Interestingly, when comparing T90 versus T0, the lipidomic profile in CGL did not change as intensely as it did for control participants. CONCLUSIONS The present study found profound alterations in the plasma lipidomic profile of complex lipids in CGL patients as compared to control subjects. A fat-containing breakfast meal did not appear to significantly influence the CGL profile observed in the fasted state. Our study may have implications for clinical practice, also aiding to a deeper comprehension of the role of complex lipids in CGL in view of novel therapeutic strategies.
Collapse
Affiliation(s)
- Camilla O D Araújo
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| | - Amanda P Pedroso
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| | - Valter T Boldarine
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| | - Anna Maria A P Fernandes
- Postgraduate Program of Health Sciences, São Francisco University, Bragança Paulista, SP, Brazil
| | - José J M Perez
- Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, SP, Brazil
| | - Renan M Montenegro
- Brazilian Group for the Study of Inherited and Acquired Lipodystrophies - Hospital Universitário Walter Cantídio, Departamento de Medicina Clínica e Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza-Ceará, Brazil
| | - Ana Paula D R Montenegro
- Brazilian Group for the Study of Inherited and Acquired Lipodystrophies - Hospital Universitário Walter Cantídio, Departamento de Medicina Clínica e Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza-Ceará, Brazil
| | - Annelise B de Carvalho
- Brazilian Group for the Study of Inherited and Acquired Lipodystrophies - Hospital Universitário Walter Cantídio, Departamento de Medicina Clínica e Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza-Ceará, Brazil
| | - Virgínia O Fernandes
- Brazilian Group for the Study of Inherited and Acquired Lipodystrophies - Hospital Universitário Walter Cantídio, Departamento de Medicina Clínica e Departamento de Saúde Comunitária, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza-Ceará, Brazil
| | - Lila M Oyama
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| | - Patrícia O Carvalho
- Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, SP, Brazil
| | - Carla S C Maia
- Departamento de Nutrição, Universidade Estadual do Ceará (UECE), Campus do Itaperi, Fortaleza, CE, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester WR2 6AJ, United Kingdom.
| | - Eliane B Ribeiro
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| | - Mônica M Telles
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, SP, Brazil
| |
Collapse
|
29
|
Guo X, Zhou Q, Jin J, Lan F, Wen C, Li J, Yang N, Sun C. Hepatic steatosis is associated with dysregulated cholesterol metabolism and altered protein acetylation dynamics in chickens. J Anim Sci Biotechnol 2023; 14:108. [PMID: 37568219 PMCID: PMC10422840 DOI: 10.1186/s40104-023-00910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/28/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Hepatic steatosis is a prevalent manifestation of fatty liver, that has detrimental effect on the health and productivity of laying hens, resulting in economic losses to the poultry industry. Here, we aimed to systematically investigate the genetic regulatory mechanisms of hepatic steatosis in laying hens. METHODS Ninety individuals with the most prominent characteristics were selected from 686 laying hens according to the accumulation of lipid droplets in the liver, and were graded into three groups, including the control, mild hepatic steatosis and severe hepatic steatosis groups. A combination of transcriptome, proteome, acetylome and lipidome analyses, along with bioinformatics analysis were used to screen the key biological processes, modifications and lipids associated with hepatic steatosis. RESULTS The rationality of the hepatic steatosis grouping was verified through liver biochemical assays and RNA-seq. Hepatic steatosis was characterized by increased lipid deposition and multiple metabolic abnormalities. Integration of proteome and acetylome revealed that differentially expressed proteins (DEPs) interacted with differentially acetylated proteins (DAPs) and were involved in maintaining the metabolic balance in the liver. Acetylation alterations mainly occurred in the progression from mild to severe hepatic steatosis, i.e., the enzymes in the fatty acid oxidation and bile acid synthesis pathways were significantly less acetylated in severe hepatic steatosis group than that in mild group (P < 0.05). Lipidomics detected a variety of sphingolipids (SPs) and glycerophospholipids (GPs) were negatively correlated with hepatic steatosis (r ≤ -0.5, P < 0.05). Furthermore, the severity of hepatic steatosis was associated with a decrease in cholesterol and bile acid synthesis and an increase in exogenous cholesterol transport. CONCLUSIONS In addition to acquiring a global and thorough picture of hepatic steatosis in laying hens, we were able to reveal the role of acetylation in hepatic steatosis and depict the changes in hepatic cholesterol metabolism. The findings provides a wealth of information to facilitate a deeper understanding of the pathophysiology of fatty liver and contributes to the development of therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoli Guo
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Qianqian Zhou
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Jiaming Jin
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Fangren Lan
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Chaoliang Wen
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Junying Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
30
|
Turk SM, Indovina CJ, Miller JM, Overton DL, Runnebohm AM, Orchard CJ, Tragesser-Tiña ME, Gosser SK, Doss EM, Richards KA, Irelan CB, Daraghmi MM, Bailey CG, Niekamp JM, Claypool KP, Engle SM, Buchanan BW, Woodruff KA, Olesen JB, Smaldino PJ, Rubenstein EM. Lipid biosynthesis perturbation impairs endoplasmic reticulum-associated degradation. J Biol Chem 2023; 299:104939. [PMID: 37331602 PMCID: PMC10372827 DOI: 10.1016/j.jbc.2023.104939] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/20/2023] Open
Abstract
The relationship between lipid homeostasis and protein homeostasis (proteostasis) is complex and remains incompletely understood. We conducted a screen for genes required for efficient degradation of Deg1-Sec62, a model aberrant translocon-associated substrate of the endoplasmic reticulum (ER) ubiquitin ligase Hrd1, in Saccharomyces cerevisiae. This screen revealed that INO4 is required for efficient Deg1-Sec62 degradation. INO4 encodes one subunit of the Ino2/Ino4 heterodimeric transcription factor, which regulates expression of genes required for lipid biosynthesis. Deg1-Sec62 degradation was also impaired by mutation of genes encoding several enzymes mediating phospholipid and sterol biosynthesis. The degradation defect in ino4Δ yeast was rescued by supplementation with metabolites whose synthesis and uptake are mediated by Ino2/Ino4 targets. Stabilization of a panel of substrates of the Hrd1 and Doa10 ER ubiquitin ligases by INO4 deletion indicates ER protein quality control is generally sensitive to perturbed lipid homeostasis. Loss of INO4 sensitized yeast to proteotoxic stress, suggesting a broad requirement for lipid homeostasis in maintaining proteostasis. A better understanding of the dynamic relationship between lipid homeostasis and proteostasis may lead to improved understanding and treatment of several human diseases associated with altered lipid biosynthesis.
Collapse
Affiliation(s)
- Samantha M Turk
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | - Jacob M Miller
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | | | - Cade J Orchard
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | | | - Ellen M Doss
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Kyle A Richards
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | | | - Connor G Bailey
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Julia M Niekamp
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | - Sarah M Engle
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | - Bryce W Buchanan
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | - James B Olesen
- Department of Biology, Ball State University, Muncie, Indiana, USA
| | | | | |
Collapse
|
31
|
Attardo GM, Benoit JB, Michalkova V, Kondragunta A, Baumann AA, Weiss BL, Malacrida A, Scolari F, Aksoy S. Lipid metabolism dysfunction following symbiont elimination is linked to altered Kennedy pathway homeostasis. iScience 2023; 26:107108. [PMID: 37534171 PMCID: PMC10391724 DOI: 10.1016/j.isci.2023.107108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/27/2023] [Accepted: 06/08/2023] [Indexed: 08/04/2023] Open
Abstract
Lipid metabolism is critical for insect reproduction, especially for species that invest heavily in the early developmental stages of their offspring. The role of symbiotic bacteria during this process is understudied but likely essential. We examined the role of lipid metabolism during the interaction between the viviparous tsetse fly (Glossina morsitans morsitans) and its obligate endosymbiotic bacteria (Wigglesworthia glossinidia) during tsetse pregnancy. We observed increased CTP:phosphocholine cytidylyltransferase (cct1) expression during pregnancy, which is critical for phosphatidylcholine biosynthesis in the Kennedy pathway. Experimental removal of Wigglesworthia impaired lipid metabolism via disruption of the Kennedy pathway, yielding obese mothers whose developing progeny starve. Functional validation via experimental cct1 suppression revealed a phenotype similar to females lacking obligate Wigglesworthia symbionts. These results indicate that, in Glossina, symbiont-derived factors, likely B vitamins, are critical for the proper function of both lipid biosynthesis and lipolysis to maintain tsetse fly fecundity.
Collapse
Affiliation(s)
- Geoffrey M. Attardo
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Entomology and Nematology, Division of Agriculture and Natural Resources, University of California Davis, Davis, CA 95616, USA
| | - Joshua B. Benoit
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Veronika Michalkova
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Section of Molecular and Applied Zoology, Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alekhya Kondragunta
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Aaron A. Baumann
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Center for Agricultural Synthetic Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Brian L. Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Anna Malacrida
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Institute of Molecular Genetics (IGM), Italian National Research Council (CNR), Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
32
|
Anari M, Montgomery MK. Phospholipid metabolism in the liver - Implications for phosphatidylserine in non-alcoholic fatty liver disease. Biochem Pharmacol 2023; 213:115621. [PMID: 37217141 DOI: 10.1016/j.bcp.2023.115621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Mammalian cells contain more than a thousand different glycerophospholipid species that are essential membrane components and signalling molecules, with phosphatidylserine (PS) giving membranes their negative surface charge. Depending on the tissue, PS is important in apoptosis, blood clotting, cancer pathogenesis, as well as muscle and brain function, processes that are dependent on the asymmetrical distribution of PS on the plasma membrane and/or the capacity of PS to act as anchorage for various signalling proteins. Recent studies have implicated hepatic PS in the progression of non-alcoholic fatty liver disease (NAFLD), either as beneficial in the context of suppressing hepatic steatosis and fibrosis, or on the other hand as a potential contributor to the progression of liver cancer. This review provides an extensive overview of hepatic phospholipid metabolism, including its biosynthetic pathways, intracellular trafficking and roles in health and disease, further taking a deeper dive into PS metabolism, including associate and causative evidence of the role of PS in advanced liver disease.
Collapse
Affiliation(s)
- Marziyeh Anari
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
33
|
Bertran L, Capellades J, Abelló S, Durán-Bertran J, Aguilar C, Martinez S, Sabench F, Correig X, Yanes O, Auguet T, Richart C. LC/MS-Based Untargeted Metabolomics Study in Women with Nonalcoholic Steatohepatitis Associated with Morbid Obesity. Int J Mol Sci 2023; 24:9789. [PMID: 37372937 DOI: 10.3390/ijms24129789] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
This study investigated the importance of a metabolomic analysis in a complex disease such as nonalcoholic steatohepatitis (NASH) associated with obesity. Using an untargeted metabolomics technique, we studied blood metabolites in 216 morbidly obese women with liver histological diagnosis. A total of 172 patients were diagnosed with nonalcoholic fatty liver disease (NAFLD), and 44 were diagnosed with normal liver (NL). Patients with NAFLD were classified into simple steatosis (n = 66) and NASH (n = 106) categories. A comparative analysis of metabolites levels between NASH and NL demonstrated significant differences in lipid metabolites and derivatives, mainly from the phospholipid group. In NASH, there were increased levels of several phosphatidylinositols and phosphatidylethanolamines, as well as isolated metabolites such as diacylglycerol 34:1, lyso-phosphatidylethanolamine 20:3 and sphingomyelin 38:1. By contrast, there were decreased levels of acylcarnitines, sphingomyelins and linoleic acid. These findings may facilitate identification studies of the main pathogenic metabolic pathways related to NASH and may also have a possible applicability in a panel of metabolites to be used as biomarkers in future algorithms of the disease diagnosis and its follow-up. Further confirmatory studies in groups with different ages and sexes are necessary.
Collapse
Affiliation(s)
- Laia Bertran
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| | - Jordi Capellades
- Department of Electronic Engineering, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Sonia Abelló
- Servei de Recursos Científics i Tècnics, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Joan Durán-Bertran
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| | - Salomé Martinez
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| | - Fàtima Sabench
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
- Unitat de Cirurgia, Facultad de Medicina i Ciències de la Salut, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
| | - Xavier Correig
- Department of Electronic Engineering, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oscar Yanes
- Department of Electronic Engineering, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, 43005 Tarragona, Spain
| |
Collapse
|
34
|
Li J, Xin Y, Li J, Chen H, Li H. Phosphatidylethanolamine N-methyltransferase: from Functions to Diseases. Aging Dis 2023; 14:879-891. [PMID: 37191416 PMCID: PMC10187709 DOI: 10.14336/ad.2022.1025] [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: 08/02/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Locating on endoplasmic reticulum and mitochondria associated membrane, Phosphatidylethanolamine N-methyltransferase (PEMT), catalyzes phosphatidylethanolamine methylation to phosphatidylcholine. As the only endogenous pathway for choline biosynthesis in mammals, the dysregulation of PEMT can lead to imbalance of phospholipid metabolism. Dysregulation of phospholipid metabolism in the liver or heart can lead to deposition of toxic lipid species that adversely result in dysfunction of hepatocyte/cardiomyocyte. Studies have shown that PEMT-/- mice increased susceptibility of diet-induced fatty liver and steatohepatitis. However, knockout of PEMT protects against diet-induced atherosclerosis, diet-induced obesity, and insulin resistance. Thus, novel insights to the function of PEMT in various organs should be summarized. Here, we reviewed the structural and functional properties of PEMT, highlighting its role in the pathogenesis of obesity, liver diseases, cardiovascular diseases, and other conditions.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hui Chen
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, China.
| |
Collapse
|
35
|
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
|
36
|
Mutation in Smek2 regulating hepatic glucose metabolism causes hypersarcosinemia and hyperhomocysteinemia in rats. Sci Rep 2023; 13:3053. [PMID: 36810603 PMCID: PMC9944932 DOI: 10.1038/s41598-022-26115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/09/2022] [Indexed: 02/24/2023] Open
Abstract
Suppressor of mek1 (Dictyostelium) homolog 2 (Smek2), was identified as one of the responsible genes for diet-induced hypercholesterolemia (DIHC) of exogenously hypercholesterolemic (ExHC) rats. A deletion mutation in Smek2 leads to DIHC via impaired glycolysis in the livers of ExHC rats. The intracellular role of Smek2 remains obscure. We used microarrays to investigate Smek2 functions with ExHC and ExHC.BN-Dihc2BN congenic rats that harbor a non-pathological Smek2 allele from Brown-Norway rats on an ExHC background. Microarray analysis revealed that Smek2 dysfunction leads to extremely low sarcosine dehydrogenase (Sardh) expression in the liver of ExHC rats. Sarcosine dehydrogenase demethylates sarcosine, a byproduct of homocysteine metabolism. The ExHC rats with dysfunctional Sardh developed hypersarcosinemia and homocysteinemia, a risk factor for atherosclerosis, with or without dietary cholesterol. The mRNA expression of Bhmt, a homocysteine metabolic enzyme and the hepatic content of betaine (trimethylglycine), a methyl donor for homocysteine methylation were low in ExHC rats. Results suggest that homocysteine metabolism rendered fragile by a shortage of betaine results in homocysteinemia, and that Smek2 dysfunction causes abnormalities in sarcosine and homocysteine metabolism.
Collapse
|
37
|
Kipp ZA, Martinez GJ, Bates EA, Maharramov AB, Flight RM, Moseley HNB, Morris AJ, Stec DE, Hinds TD. Bilirubin Nanoparticle Treatment in Obese Mice Inhibits Hepatic Ceramide Production and Remodels Liver Fat Content. Metabolites 2023; 13:215. [PMID: 36837834 PMCID: PMC9965094 DOI: 10.3390/metabo13020215] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Studies have indicated that increasing plasma bilirubin levels might be useful for preventing and treating hepatic lipid accumulation that occurs with metabolic diseases such as obesity and diabetes. We have previously demonstrated that mice with hyperbilirubinemia had significantly less lipid accumulation in a diet-induced non-alcoholic fatty liver disease (NAFLD) model. However, bilirubin's effects on individual lipid species are currently unknown. Therefore, we used liquid chromatography-mass spectroscopy (LC-MS) to determine the hepatic lipid composition of obese mice with NAFLD treated with bilirubin nanoparticles or vehicle control. We placed the mice on a high-fat diet (HFD) for 24 weeks and then treated them with bilirubin nanoparticles or vehicle control for 4 weeks while maintaining the HFD. Bilirubin nanoparticles suppressed hepatic fat content overall. After analyzing the lipidomics data, we determined that bilirubin inhibited the accumulation of ceramides in the liver. The bilirubin nanoparticles significantly lowered the hepatic expression of two essential enzymes that regulate ceramide production, Sgpl1 and Degs1. Our results demonstrate that the bilirubin nanoparticles improve hepatic fat content by reducing ceramide production, remodeling the liver fat content, and improving overall metabolic health.
Collapse
Affiliation(s)
- Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Evelyn A Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Agil B Maharramov
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
| | - Robert M Flight
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Hunter N B Moseley
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
- Institute for Biomedical Informatics, University of Kentucky, Lexington, KY 40508, USA
- Center for Clinical and Translational Sciences, University of Kentucky, Lexington, KY 40508, USA
| | - Andrew J Morris
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - David E Stec
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 760 Press Avenue, Healthy Kentucky Research Building, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
| |
Collapse
|
38
|
Shama S, Jang H, Wang X, Zhang Y, Shahin NN, Motawi TK, Kim S, Gawrieh S, Liu W. Phosphatidylethanolamines Are Associated with Nonalcoholic Fatty Liver Disease (NAFLD) in Obese Adults and Induce Liver Cell Metabolic Perturbations and Hepatic Stellate Cell Activation. Int J Mol Sci 2023; 24:ijms24021034. [PMID: 36674549 PMCID: PMC9861886 DOI: 10.3390/ijms24021034] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Pathogenesis roles of phospholipids (PLs) in nonalcoholic fatty liver disease (NAFLD) remain incompletely understood. This study investigated the role of PLs in the progression of NAFLD among obese individuals via studying the alterations in serum PL composition throughout the spectrum of disease progression and evaluating the effects of specific phosphatidylethanolamines (PEs) on FLD development in vitro. A total of 203 obese subjects, who were undergoing bariatric surgery, were included in this study. They were histologically classified into 80 controls (C) with normal liver histology, 93 patients with simple hepatic steatosis (SS), 16 with borderline nonalcoholic steatohepatitis (B-NASH) and 14 with progressive NASH (NASH). Serum PLs were profiled by automated electrospray ionization tandem mass spectrometry (ESI-MS/MS). HepG2 (hepatoma cells) and LX2 (immortalized hepatic stellate cells or HSCs) were used to explore the roles of PL in NAFLD/NASH development. Several PLs and their relative ratios were significantly associated with NAFLD progression, especially those involving PE. Incubation of HepG2 cells with two phosphatidylethanolamines (PEs), PE (34:1) and PE (36:2), resulted in significant inhibition of cell proliferation, reduction of mitochondrial mass and membrane potential, induction of lipid accumulation and mitochondrial ROS production. Meanwhile, treatment of LX2 cells with both PEs markedly increased cell activation and migration. These effects were associated with a significant change in the expression levels of genes involved in lipogenesis, lipid oxidation, autophagy, apoptosis, inflammation, and fibrosis. Thus, our study demonstrated that elevated level of PEs increases susceptibility to the disease progression of obesity associated NAFLD, likely through a causal cascade of impacts on the function of different liver cells.
Collapse
Affiliation(s)
- Samaa Shama
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Cell-Based Analysis Unit, Reference Laboratory, Egyptian Drug Authority, Cairo 12618, Egypt
| | - Hyejeong Jang
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Yang Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Tarek Kamal Motawi
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| | - Seongho Kim
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Samer Gawrieh
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence: (T.K.M.); (W.L.); Tel.: +20-122-313-8667 (T.K.M.); +1-313-577-3375 (W.L.)
| |
Collapse
|
39
|
Javaid A, Wang F, Horst EA, Diaz-Rubio ME, Wang LF, Baumgard LH, McFadden JW. Effects of acute intravenous lipopolysaccharide administration on the plasma lipidome and metabolome in lactating Holstein cows experiencing hyperlipidemia. Metabolomics 2022; 18:75. [PMID: 36125563 DOI: 10.1007/s11306-022-01928-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The effects of lipopolysaccharides (i.e., endotoxin; LPS) on metabolism are poorly defined in lactating dairy cattle experiencing hyperlipidemia. OBJECTIVES Our objective was to explore the effects of acute intravenous LPS administration on metabolism in late-lactation Holstein cows experiencing hyperlipidemia induced by intravenous triglyceride infusion and feed restriction. METHODS Ten non-pregnant lactating Holstein cows (273 ± 35 d in milk) were administered a single bolus of saline (3 mL of saline; n [Formula: see text] 5) or LPS (0.375 [Formula: see text]g of LPS/kg of body weight; n [Formula: see text] 5). Simultaneously, cows were intravenously infused a triglyceride emulsion and feed restricted for 16 h to induce hyperlipidemia in an attempt to model the periparturient period. Blood was sampled at routine intervals. Changes in circulating total fatty acid concentrations and inflammatory parameters were measured. Plasma samples were analyzed using untargeted lipidomics and metabolomics. RESULTS Endotoxin increased circulating serum amyloid A, LPS-binding protein, and cortisol concentrations. Endotoxin administration decreased plasma lysophosphatidylcholine (LPC) concentrations and increased select plasma ceramide concentrations. These outcomes suggest modulation of the immune response and insulin action. Lipopolysaccharide decreased the ratio of phosphatidylcholine to phosphatidylethanomanine, which potentially indicate a decrease in the hepatic activation of phosphatidylethanolamine N-methyltransferase and triglyceride export. Endotoxin administration also increased plasma concentrations of pyruvic and lactic acids, and decreased plasma citric acid concentrations, which implicate the upregulation of glycolysis and downregulation of the citric acid cycle (i.e., the Warburg effect), potentially in leukocytes. CONCLUSION Acute intravenous LPS administration decreased circulating LPC concentrations, modified ceramide and glycerophospholipid concentrations, and influenced intermediary metabolism in dairy cows experiencing hyperlipidemia.
Collapse
Affiliation(s)
- Awais Javaid
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA.
| | - Feiran Wang
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
- China Agricultural University, Beijing, 100193, China
| | - Erin A Horst
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - M Elena Diaz-Rubio
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, 14853, USA
| | - Lin F Wang
- Henan Agricultural University, Zhengzhou, 450002, China
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Joseph W McFadden
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
40
|
Zhang C, Shi M, Kim W, Arif M, Klevstig M, Li X, Yang H, Bayram C, Bolat I, Tozlu ÖÖ, Hacımuftuoglu A, Yıldırım S, Sebhaoui J, Iqbal S, Wei Y, Shi X, Nielsen J, Turkez H, Uhlen M, Boren J, Mardinoglu A. Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositioning. EBioMedicine 2022; 83:104214. [PMID: 35988463 PMCID: PMC9420484 DOI: 10.1016/j.ebiom.2022.104214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/22/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) encompasses a wide spectrum of liver pathologies. However, no medical treatment has been approved for the treatment of NAFLD. In our previous study, we found that PKLR could be a potential target for treatment of NALFD. Here, we investigated the effect of PKLR in in vivo model and performed drug repositioning to identify a drug candidate for treatment of NAFLD. METHODS Tissue samples from liver, muscle, white adipose and heart were obtained from control and PKLR knockout mice fed with chow and high sucrose diets. Lipidomics as well as transcriptomics analyses were conducted using these tissue samples. In addition, a computational drug repositioning analysis was performed and drug candidates were identified. The drug candidates were both tested in in vitro and in vivo models to evaluate their toxicity and efficacy. FINDINGS The Pklr KO reversed the increased hepatic triglyceride level in mice fed with high sucrose diet and partly recovered the transcriptomic changes in the liver as well as in other three tissues. Both liver and white adipose tissues exhibited dysregulated circadian transcriptomic profiles, and these dysregulations were reversed by hepatic knockout of Pklr. In addition, 10 small molecule drug candidates were identified as potential inhibitor of PKLR using our drug repositioning pipeline, and two of them significantly inhibited both the PKLR expression and triglyceride level in in vitro model. Finally, the two selected small molecule drugs were evaluated in in vivo rat models and we found that these drugs attenuate the hepatic steatosis without side effect on other tissues. INTERPRETATION In conclusion, our study provided biological insights about the critical role of PKLR in NAFLD progression and proposed a treatment strategy for NAFLD patients, which has been validated in preclinical studies. FUNDING ScandiEdge Therapeutics and Knut and Alice Wallenberg Foundation.
Collapse
Affiliation(s)
- Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Mengnan Shi
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Martina Klevstig
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Hong Yang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, 25240, Erzurum, Turkey
| | - Ismail Bolat
- Department of Pathology, Veterinary Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Özlem Özdemir Tozlu
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25200 Erzurum, Turkey
| | - Ahmet Hacımuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, 25240, Erzurum, Turkey
| | - Serkan Yıldırım
- Department of Pathology, Veterinary Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Jihad Sebhaoui
- Trustlife Laboratories, Drug Research & Development Center, Istanbul, Turkey
| | - Shazia Iqbal
- Trustlife Laboratories, Drug Research & Development Center, Istanbul, Turkey
| | - Yongjun Wei
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Xiaojing Shi
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan Province, 450001, PR China
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
41
|
Stork BA, Dean A, Ortiz AR, Saha P, Putluri N, Planas-Silva MD, Mahmud I, Rajapakshe K, Coarfa C, Knapp S, Lorenzi PL, Kemp BE, Turk BE, Scott JW, Means AR, York B. Calcium/calmodulin-dependent protein kinase kinase 2 regulates hepatic fuel metabolism. Mol Metab 2022; 62:101513. [PMID: 35562082 PMCID: PMC9157561 DOI: 10.1016/j.molmet.2022.101513] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The liver is the primary internal metabolic organ that coordinates whole body energy homeostasis in response to feeding and fasting. Genetic ablation or pharmacological inhibition of calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) has been shown to significantly improve hepatic health and peripheral insulin sensitivity upon overnutrition with high fat diet. However, the precise molecular underpinnings that explain this metabolic protection have remained largely undefined. METHODS To characterize the role of CaMKK2 in hepatic metabolism, we developed and challenged liver-specific CaMKK2 knockout (CaMKK2LKO) mice with high fat diet and performed glucose and insulin tolerance tests to evaluate peripheral insulin sensitivity. We used a combination of RNA-Sequencing, glucose and fatty acid istotopic tracer studies, a newly developed Seahorse assay for measuring the oxidative capacity of purified peroxisomes, and a degenerate peptide libarary to identify putative CaMKK2 substrates that mechanistically explain the protective effects of hepatic CaMKK2 ablation. RESULTS Consistent with previous findings, we show that hepatic CaMKK2 ablation significantly improves indices of peripheral insulin sensitivity. Mechanistically, we found that CaMKK2 phosphorylates and regulates GAPDH to promote glucose metabolism and PEX3 to blunt peroxisomal fatty acid catabolism in the liver. CONCLUSION CaMKK2 is a central metabolic fuel sensor in the liver that significantly contributes to whole body systems metabolism.
Collapse
Affiliation(s)
- Brittany A Stork
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Adam Dean
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrea R Ortiz
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pradip Saha
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nagireddy Putluri
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Iqbal Mahmud
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kimal Rajapakshe
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stefan Knapp
- Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Victoria, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Benjamin E Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - John W Scott
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Victoria, 3065, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Brian York
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
42
|
Zhang LY, Shi HH, Wang CC, Wang YM, Wei ZH, Xue CH, Mao XZ, Zhang TT. Targeted Lipidomics Reveal the Effects of Different Phospholipids on the Phospholipid Profiles of Hepatic Mitochondria and Endoplasmic Reticulum in High-Fat/High-Fructose-Diet-Induced Nonalcoholic Fatty Liver Disease Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3529-3540. [PMID: 35212227 DOI: 10.1021/acs.jafc.1c07538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The lipid alternation in mitochondria and endoplasmic reticulum (ER) might be indicative of their abnormal morphology and function, which contribute to development of nonalcoholic fatty liver disease (NAFLD). However, the influence of dietary phospholipids (PLs) on the PL composition of the organellar membrane is largely unknown. High-fat/high-fructose (HFHF)-diet-induced NAFLD mice were administrated with different PLs (2%, w/w) with specific fatty acids and headgroups, including eicosapentaenoic acid (EPA)-phosphatidylcholine (PC)/phosphatidylethanolamine (PE)/phosphatidylserine (PS), docosahexaenoic acid (DHA)-PC/PE/PS, egg-PC/PE/PS, and soy-PC/PE/PS. After 8 weeks of feeding, PLs dramatically decreased hepatic lipid accumulation, in which EPA/DHA-PS had the best efficiency. Furthermore, lipidomic analysis revealed that the HFHF diet narrowed the difference in PL composition between mitochondria and ER, significantly reduced the PC/PE ratio, and changed the unsaturation of cardiolipin in mitochondria. Dietary PLs reversed these alterations. Heatmap analysis indicated that dietary PL groups containing the same fatty acids clustered together. Moreover, dietary PLs significantly increased the ratio of PC/PE in both hepatic mitochondria and ER, especially EPA-PE. This study showed that fatty acid composition of PLs might represent greater impact on the PL composition of the organellar membrane than headgroups.
Collapse
Affiliation(s)
- Ling-Yu Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, People's Republic of China
| | - Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| | - Zi-Hao Wei
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| | - Xiang-Zhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| |
Collapse
|
43
|
Di Ciaula A, Bonfrate L, Krawczyk M, Frühbeck G, Portincasa P. Synergistic and Detrimental Effects of Alcohol Intake on Progression of Liver Steatosis. Int J Mol Sci 2022; 23:2636. [PMID: 35269779 PMCID: PMC8910376 DOI: 10.3390/ijms23052636] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are the most common liver disorders worldwide and the major causes of non-viral liver cirrhosis in the general population. In NAFLD, metabolic abnormalities, obesity, and metabolic syndrome are the driving factors for liver damage with no or minimal alcohol consumption. ALD refers to liver damage caused by excess alcohol intake in individuals drinking more than 5 to 10 daily units for years. Although NAFLD and ALD are nosologically considered two distinct entities, they show a continuum and exert synergistic effects on the progression toward liver cirrhosis. The current view is that low alcohol use might also increase the risk of advanced clinical liver disease in NAFLD, whereas metabolic factors increase the risk of cirrhosis among alcohol risk drinkers. Therefore, special interest is now addressed to individuals with metabolic abnormalities who consume small amounts of alcohol or who binge drink, for the role of light-to-moderate alcohol use in fibrosis progression and clinical severity of the liver disease. Evidence shows that in the presence of NAFLD, there is no liver-safe limit of alcohol intake. We discuss the epidemiological and clinical features of NAFLD/ALD, aspects of alcohol metabolism, and mechanisms of damage concerning steatosis, fibrosis, cumulative effects, and deleterious consequences which include hepatocellular carcinoma.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
| | - Leonilde Bonfrate
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
| | - Marcin Krawczyk
- Department of Medicine II Saarland University Medical Center, Saarland University, 66424 Homburg, Germany;
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31009 Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31009 Pamplona, Spain
| | - Piero Portincasa
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
| |
Collapse
|
44
|
Aragón-Herrera A, Otero-Santiago M, Anido-Varela L, Moraña-Fernández S, Campos-Toimil M, García-Caballero T, Barral L, Tarazón E, Roselló-Lletí E, Portolés M, Gualillo O, Moscoso I, Lage R, González-Juanatey JR, Feijóo-Bandín S, Lago F. The Treatment With the SGLT2 Inhibitor Empagliflozin Modifies the Hepatic Metabolome of Male Zucker Diabetic Fatty Rats Towards a Protective Profile. Front Pharmacol 2022; 13:827033. [PMID: 35185578 PMCID: PMC8847595 DOI: 10.3389/fphar.2022.827033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/04/2022] [Indexed: 12/22/2022] Open
Abstract
The EMPA-REG OUTCOME (Empagliflozin, Cardiovascular Outcome Event Trial in patients with Type 2 Diabetes Mellitus (T2DM)) trial evidenced the potential of sodium-glucose cotransporter 2 (SGLT2) inhibitors for the treatment of patients with diabetes and cardiovascular disease. Recent evidences have shown the benefits of the SGLT2 inhibitor empagliflozin on improving liver steatosis and fibrosis in patients with T2DM. Metabolomic studies have been shown to be very useful to improve the understanding of liver pathophysiology during the development and progression of metabolic hepatic diseases, and because the effects of empagliflozin and of other SGLT2 inhibitors on the complete metabolic profile of the liver has never been analysed before, we decided to study the impact on the liver of male Zucker diabetic fatty (ZDF) rats of a treatment for 6 weeks with empagliflozin using an untargeted metabolomics approach, with the purpose to help to clarify the benefits of the use of empagliflozin at hepatic level. We found that empagliflozin is able to change the hepatic lipidome towards a protective profile, through an increase of monounsaturated and polyunsaturated glycerides, phosphatidylcholines, phosphatidylethanolamines, lysophosphatidylinositols and lysophosphatidylcholines. Empagliflozin also induces a decrease in the levels of the markers of inflammation IL-6, chemerin and chemerin receptor in the liver. Our results provide new evidences regarding the molecular pathways through which empagliflozin could exert hepatoprotector beneficial effects in T2DM.
Collapse
Affiliation(s)
- Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Manuel Otero-Santiago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Laura Anido-Varela
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Sandra Moraña-Fernández
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Manuel Campos-Toimil
- Group of Pharmacology of Chronic Diseases (CD Pharma), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tomás García-Caballero
- Department of Morphological Sciences, University of Santiago de Compostela and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Luis Barral
- Group of Polymers, Department of Physics and Earth Sciences, University of La Coruña, La Coruña, Spain
| | - Estefanía Tarazón
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain.,Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Esther Roselló-Lletí
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain.,Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Manuel Portolés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain.,Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Valencia, Spain
| | - Oreste Gualillo
- Laboratory of Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Isabel Moscoso
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) and Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ricardo Lage
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) and Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Diets Rich in Olive Oil, Palm Oil, or Lard Alter Mitochondrial Biogenesis and Mitochondrial Membrane Composition in Rat Liver. Biochem Res Int 2022; 2022:9394356. [PMID: 35237451 PMCID: PMC8885195 DOI: 10.1155/2022/9394356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/16/2022] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
Palm oil (crude or refined) and lard are rich in SFA, while olive oil is rich in polyunsaturated fatty acids. SFA are considered harmful to health, while polyunsaturated fatty acids are beneficial to health. The aim of this study was to determine the effect of diets rich in crude PO, refined PO, OO, or lard on the mitochondrial membrane, the activity of mitochondrial respiratory chain complexes, and mitochondrial biogenesis. This was an experimental study in male Wistar rats fed a diet containing 30% of each oil. Rats had free access to food and water. After being fed for 12 weeks, animals were sacrificed and liver mitochondria were collected. This collection was used to determine membrane potential and ROS production, membrane phospholipid and fatty acid composition, citrate synthase activity and respiratory chain complex, cardiolipin synthase protein expression, and expression of selected genes involved in mitochondrial biogenesis. We found that diets rich in olive oil, palm oil, or lard altered mitochondrial biogenesis by significantly decreasing Pgc1α gene expression and altered the fatty acid composition of rat liver mitochondrial membrane PL.
Collapse
|
46
|
Martinou E, Pericleous M, Stefanova I, Kaur V, Angelidi AM. Diagnostic Modalities of Non-Alcoholic Fatty Liver Disease: From Biochemical Biomarkers to Multi-Omics Non-Invasive Approaches. Diagnostics (Basel) 2022; 12:407. [PMID: 35204498 PMCID: PMC8871470 DOI: 10.3390/diagnostics12020407] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is currently the most common cause of chronic liver disease worldwide, and its prevalence is increasing globally. NAFLD is a multifaceted disorder, and its spectrum includes steatosis to steatohepatitis, which may evolve to advanced fibrosis and cirrhosis. In addition, the presence of NAFLD is independently associated with a higher cardiometabolic risk and increased mortality rates. Considering that the vast majority of individuals with NAFLD are mainly asymptomatic, early diagnosis of non-alcoholic steatohepatitis (NASH) and accurate staging of fibrosis risk is crucial for better stratification, monitoring and targeted management of patients at risk. To date, liver biopsy remains the gold standard procedure for the diagnosis of NASH and staging of NAFLD. However, due to its invasive nature, research on non-invasive tests is rapidly increasing with significant advances having been achieved during the last decades in the diagnostic field. New promising non-invasive biomarkers and techniques have been developed, evaluated and assessed, including biochemical markers, imaging modalities and the most recent multi-omics approaches. Our article provides a comprehensive review of the currently available and emerging non-invasive diagnostic tools used in assessing NAFLD, also highlighting the importance of accurate and validated diagnostic tools.
Collapse
Affiliation(s)
- Eirini Martinou
- Hepatobiliary and Pancreatic Surgery Department, Royal Surrey County Hospital, Guildford GU2 7XX, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK;
| | - Marinos Pericleous
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK;
- Department of Gastroenterology and Hepatology, Royal Surrey County Hospital, Guildford GU2 7XX, UK
| | - Irena Stefanova
- Department of General Surgery, Frimley Health NHS Foundation Trust, Camberley GU16 7UJ, UK;
| | - Vasha Kaur
- Department of Upper Gastrointestinal and Bariatric Surgery, St George’s Hospital, London SW17 0QT, UK;
| | - Angeliki M. Angelidi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
47
|
Li H, Xu QY, Xie Y, Luo JJ, Cao HX, Pan Q. Effects of chronic HBV infection on lipid metabolism in non-alcoholic fatty liver disease: A lipidomic analysis. Ann Hepatol 2022; 24:100316. [PMID: 33515803 DOI: 10.1016/j.aohep.2021.100316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Chronic hepatitis B virus (HBV) infection exerts an impact on lipid metabolism, but its interaction with dysmetabolism-based non-alcoholic fatty liver disease (NAFLD) remains uncertain. The purpose of the study was to investigate the effects of HBV infection on lipid metabolism, hepatic steatosis and related impairments of NAFLD patients. METHODS Biopsy-proven Chinese NAFLD patients with (NAFLD-HBV group, n = 21) or without chronic HBV infection (NAFLD group, n = 41) were enrolled in the case-control study. Their serum lipidomics was subjected to individual investigation by ultra-performance liquid chromatography-tandem mass spectrometry. Steatosis, activity, and fibrosis (SAF) scoring revealed the NAFLD-specific pathological characteristics. RESULTS Chronic HBV infection was associated with global alteration of serum lipidomics in NAFLD patients. Upregulation of phosphatidylcholine (PCs), choline plasmalogen (PC-Os) and downregulation of free fatty acids (FFAs), lysophosphatidylcholine (LPCs) dominated the HBV-related lipidomic characteristics. Compared to those of NAFLD group, the levels of serum hepatoxic lipids (FFA16:0, FFA16: 1, FFA18:1, FFA18:2) were significantly lowered in the NAFLD-HBV group. These low-level FFAs demonstrated correlation to statistical improvements in aspartate aminotransferase activity (FFA16:0, r = 0.33; FFA16:1, r = 0.37; FFA18:1, r = 0.32; FFA18:2, r = 0.42), hepatocyte steatosis (FFA16: 1, r = 0.39; FFA18:1, r = 0.39; FFA18:2, r = 0.32), and ballooning (FFA16:0, r = 0.30; FFA16:1, r = 0.45; FFA18:1, r = 0.36; FFA18:2, r = 0.30) (all P < 0.05). CONCLUSION Chronic HBV infection may impact on the serum lipidomics and steatosis-related pathological characteristics of NAFLD.
Collapse
Affiliation(s)
- Han Li
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qing-Yang Xu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yang Xie
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ji-Jun Luo
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hai-Xia Cao
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Qin Pan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
48
|
Zhang L, Hu Y, An Y, Wang Q, Liu J, Wang G. The Changes of Lipidomic Profiles Reveal Therapeutic Effects of Exenatide in Patients With Type 2 Diabetes. Front Endocrinol (Lausanne) 2022; 13:677202. [PMID: 35432194 PMCID: PMC9009038 DOI: 10.3389/fendo.2022.677202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Exenatide has been demonstrated beneficial effects on patients with type 2 diabetes mellitus (T2DM) regarding lipid metabolism. However, the potential mechanism remains unclear. We used a lipidomic approach to evaluate lipid changes in response to treatment with exenatide in T2DM patients. METHODS Serum lipidomic profiles of 35 newly diagnosed T2DM patients (before and after exenatide treatment) and 20 age-matched healthy controls were analyzed by ultrahigh-performance liquid chromatography-tandem quadrupole time-of-flight mass spectrometry. RESULTS A total of 45 lipid species including sphingomyelins (SMs), ceramides (CERs), lysophosphatidylcholines (LPCs), phosphatidylethanolamines (PEs), lysophosphatidylethanolamines (LPEs) and phosphatidylcholines (PCs) were identified in all participants. Compared to the healthy controls, 13 lipid species [SM (d18:1/18:0, d18:1/18:1), Cer (d18:1/18:0, d18:1/16:0, d18:1/20:0, d18:1/24:1), LPC (15:0, 16:0, 17:0), PC (19:0/19:0), LPE (18:0) and PE (16:0/22:6, 18:0/22:6)] were markedly increased in the T2DM group, while PE (17:0/17:0) and PC (18:1/18:0) were decreased (P < 0.05). The serum SM (d18:1/18:0, d18:1/18:1), LPC (16:0), and LPE (18:0) were significantly decreased after exenatide treatment, which was accompanied by the amelioration of lipids and glycemic parameters (TC, LDL-C, ApoA-I, FCP and HbA1c) in T2DM patients. The chord diagrams showed distinct correlation patterns between lipid classes and subclasses among healthy controls, T2DM patients before and after exenatide treatment. CONCLUSION Our results revealed that the therapeutic benefits of exenatide on T2DM might be involved in the improved lipid metabolism, especially SM, LPC, and LPE. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT03297879.
Collapse
Affiliation(s)
| | | | | | | | - Jia Liu
- *Correspondence: Jia Liu, ; Guang Wang,
| | | |
Collapse
|
49
|
Zhang S, Hu L, Han C, Huang R, Ooi K, Qian X, Ren X, Chu D, Zhang H, Du D, Xia C. PLIN2 Mediates Neuroinflammation and Oxidative/Nitrosative Stress via Downregulating Phosphatidylethanolamine in the Rostral Ventrolateral Medulla of Stressed Hypertensive Rats. J Inflamm Res 2021; 14:6331-6348. [PMID: 34880641 PMCID: PMC8646230 DOI: 10.2147/jir.s329230] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/03/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose Oxidative/nitrosative stress, neuroinflammation and their intimate interactions mediate sympathetic overactivation in hypertension. An immoderate inflammatory response is characterized not only by elevated proinflammatory cytokines (PICs) but by increases in mitochondrial dysfunction, reactive oxygen species (ROS), and nitric oxide (NO). Recent data pinpoint that both the phospholipid and lipid droplets (LDs) are potent modulators of microglia physiology. Methods Stress rats underwent compound stressors for 15 days with PLIN2-siRNA or scrambled-siRNA (SC-siRNA) administrated into the rostral ventrolateral medulla (RVLM). Lipids were analyzed by mass spectroscopy-based quantitative lipidomics. The phenotypes and proliferation of microglia, LDs, in the RVLM of rats were detected; blood pressure (BP) and myocardial injury in rats were evaluated. The anti-oxidative/nitrosative stress effect of phosphatidylethanolamine (PE) was explored in cultured primary microglia. Results Lipidomics analysis showed that 75 individual lipids in RVLM were significantly dysregulated by stress [PE was the most one], demonstrating that lipid composition changed with stress. In vitro, prorenin stress induced the accumulation of LDs, increased PICs, which could be blocked by siRNA-PLIN2 in microglia. PLIN2 knockdown upregulated the PE synthesis in microglia. Anti-oxidative/nitrosative stress effect of PE delivery was confirmed by the decrease of ROS and decrease in 3-NT and MDA in prorenin-treated microglia. PLIN2 knockdown in the RVLM blocked the number of iNOS+ and PCNA+ microglia, decreased BP, alleviated cardiac fibrosis and hypertrophy in stressed rats. Conclusion PLIN2 mediates microglial polarization/proliferation via downregulating PE in the RVLM of stressed rats. Delivery of PE is a promising strategy for combating neuroinflammation and oxidative/nitrosative stress in stress-induced hypertension.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Li Hu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200240, People's Republic of China
| | - Chengzhi Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Renhui Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Kokwin Ooi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xinyi Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaorong Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dechang Chu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, People's Republic of China
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, People's Republic of China
| | - Dongshu Du
- School of Life Science, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
50
|
Papadopoulos C, Tentes I, Anagnostopoulos K. Lipotoxicity Disrupts Erythrocyte Function: A Perspective. Cardiovasc Hematol Disord Drug Targets 2021; 21:91-94. [PMID: 34825642 DOI: 10.2174/1871529x21666210719125728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/25/2021] [Accepted: 03/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lipid accumulation in the liver, skeletal and cardiac muscle, kidneys and pancreas causes cell dysfunction, death and inflammation, a biological phenomenon named lipotoxicity. Erythrocytes participate in the transport of lipids in the circulation, and their lipidome is determined by exchange with blood components. OBJECTIVE The objective of this study is to summarize the current knowledge regarding the effect of toxic lipid accumulation in erythrocytes. RESULTS Erythrocyte lipidome is altered in lipotoxic diseases, such as fatty liver disease, heart failure and diabetes. In addition, ceramide, lysophosphatidylcholine, lysophosphatidic acid, palmitic acid and free cholesterol induce erythrocyte malfunction. CONCLUSION Erythrocytes are an additional cell target of lipotoxicity. Further exploration of the implicated molecular mechanisms could lead to novel therapeutic targets for cardiometabolic and hematological diseases.
Collapse
Affiliation(s)
- Charalampos Papadopoulos
- Laboratory of Biochemistry, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Tentes
- Laboratory of Biochemistry, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|