1
|
Zheng D, Jin S, Liu PS, Ye J, Xie X. Targeting ferroptosis by natural products in pathophysiological conditions. Arch Toxicol 2024; 98:3191-3208. [PMID: 38987487 DOI: 10.1007/s00204-024-03812-4] [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: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Ferroptosis is a form of cell death that is induced by iron-mediated accumulation of lipid peroxidation. The involvement of ferroptosis in different pathophysiological conditions has offered new perspectives on potential therapeutic interventions. Natural products, which are widely recognized for their significance in drug discovery and repurposing, have shown great promise in regulating ferroptosis by targeting various ferroptosis players. In this review, we discuss the regulatory mechanisms of ferroptosis and its implications in different pathological conditions. We dissect the interactions between natural products and ferroptosis in cancer, ischemia/reperfusion, neurodegenerative diseases, acute kidney injury, liver injury, and cardiomyopathy, with an emphasis on the relevance of ferroptosis players to disease targetability.
Collapse
Affiliation(s)
- Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jianping Ye
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
2
|
Peleman C, Francque S, Berghe TV. Emerging role of ferroptosis in metabolic dysfunction-associated steatotic liver disease: revisiting hepatic lipid peroxidation. EBioMedicine 2024; 102:105088. [PMID: 38537604 PMCID: PMC11026979 DOI: 10.1016/j.ebiom.2024.105088] [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: 12/31/2023] [Revised: 02/22/2024] [Accepted: 03/12/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is characterised by cell death of parenchymal liver cells which interact with their microenvironment to drive disease activity and liver fibrosis. The identification of the major death type could pave the way towards pharmacotherapy for MASH. To date, increasing evidence suggest a type of regulated cell death, named ferroptosis, which occurs through iron-catalysed peroxidation of polyunsaturated fatty acids (PUFA) in membrane phospholipids. Lipid peroxidation enjoys renewed interest in the light of ferroptosis, as druggable target in MASH. This review recapitulates the molecular mechanisms of ferroptosis in liver physiology, evidence for ferroptosis in human MASH and critically appraises the results of ferroptosis targeting in preclinical MASH models. Rewiring of redox, iron and PUFA metabolism in MASH creates a proferroptotic environment involved in MASH-related hepatocellular carcinoma (HCC) development. Ferroptosis induction might be a promising novel approach to eradicate HCC, while its inhibition might ameliorate MASH disease progression.
Collapse
Affiliation(s)
- Cédric Peleman
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Sven Francque
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium.
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Tao Q, Liu N, Wu J, Chen J, Chen X, Peng C. Mefloquine enhances the efficacy of anti-PD-1 immunotherapy via IFN-γ-STAT1-IRF1-LPCAT3-induced ferroptosis in tumors. J Immunother Cancer 2024; 12:e008554. [PMID: 38471712 DOI: 10.1136/jitc-2023-008554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Ferroptosis plays an important role in enhancing the efficacy of anti-programmed cell death 1 (PD-1) immunotherapy; however, the molecular mechanisms by which tumor ferroptosis sensitizes melanoma and lung cancer to anti-PD-1 immunotherapy have not been elucidated. METHODS Cytotoxicity assays, colony formation assays, flow cytometry and animal experiments were used to evaluate the effects of mefloquine (Mef) on survival and ferroptosis in melanoma and lung cancer. RNA sequencing, Real-time quantitative PCR (qRT-PCR), western blotting, chromatin immunoprecipitation-qPCR and flow cytometry were used to determine the molecular mechanisms by which Mef regulates lysophosphatidylcholine acyltransferase 3 (LPCAT3). The relationship between LPCAT3 and the efficacy of anti-PD-1 immunotherapy was verified via a clinical database and single-cell RNA sequencing (ScRNA-Seq). RESULTS In this study, we discovered that Mef induces ferroptosis. Furthermore, treatment with Mef in combination with T-cell-derived interferon-γ (IFN-γ) enhanced tumor ferroptosis and sensitized melanoma and lung cancer cells to anti-PD-1 immunotherapy. Mechanistically, Mef upregulated the expression of LPCAT3, a key gene involved in lipid peroxidation, by activating IFN-γ-induced STAT1-IRF1 signaling, and knocking down LPCAT3 impaired the induction of ferroptosis by Mef+IFN-γ. Clinically, analysis of the transcriptome and single-cell sequencing results in patients with melanoma showed that LPCAT3 expression was significantly lower in patients with melanoma than in control individuals, and LPCAT3 expression was positively correlated with the efficacy of anti-PD-1 immunotherapy. CONCLUSIONS In conclusion, our study demonstrated a novel mechanism by which LPCAT3 is regulated, and demonstrated that Mef is a highly promising new target that can be utilized to enhance the efficacy of anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Qian Tao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nian Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Jin S, Liu PS, Zheng D, Xie X. The interplay of miRNAs and ferroptosis in diseases related to iron overload. Apoptosis 2024; 29:45-65. [PMID: 37758940 DOI: 10.1007/s10495-023-01890-w] [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] [Accepted: 09/01/2023] [Indexed: 09/29/2023]
Abstract
Ferroptosis has been conceptualized as a novel cell death modality distinct from apoptosis, necroptosis, pyroptosis and autophagic cell death. The sensitivity of cellular ferroptosis is regulated at multiple layers, including polyunsaturated fatty acid metabolism, glutathione-GPX4 axis, iron homeostasis, mitochondria and other parallel pathways. In addition, microRNAs (miRNAs) have been implicated in modulating ferroptosis susceptibility through targeting different players involved in the execution or avoidance of ferroptosis. A growing body of evidence pinpoints the deregulation of miRNA-regulated ferroptosis as a critical factor in the development and progression of various pathophysiological conditions related to iron overload. The revelation of mechanisms of miRNA-dependent ferroptosis provides novel insights into the etiology of diseases and offers opportunities for therapeutic intervention. In this review, we discuss the interplay of emerging miRNA regulators and ferroptosis players under different pathological conditions, such as cancers, ischemia/reperfusion, neurodegenerative diseases, acute kidney injury and cardiomyopathy. We emphasize on the relevance of miRNA-regulated ferroptosis to disease progression and the targetability for therapeutic interventions.
Collapse
Affiliation(s)
- Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Pu-Ste Liu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan, ROC
| | - Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China.
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China.
| |
Collapse
|
5
|
Chen L, Xue J, Zhao L, He Y, Fu S, Ma X, Yu W, Tang Y, Wang Y, Gao Z. Lysophosphatidylcholine acyltransferase level predicts the severity and prognosis of patients with community-acquired pneumonia: a prospective multicenter study. Front Immunol 2024; 14:1295353. [PMID: 38259459 PMCID: PMC10800399 DOI: 10.3389/fimmu.2023.1295353] [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: 09/16/2023] [Accepted: 12/07/2023] [Indexed: 01/24/2024] Open
Abstract
Background Identifying the diagnosis as well as prognosis for patients presented with community-acquired pneumonia (CAP) remains challenging. We aimed to identify the role of lysophosphatidylcholine acyl-transferase (LPCAT) for CAP along with assessing this protein's effectiveness as a biomarker for severity of disease and mortality. Methods Prospective multicenter research study was carried out among hospitalized patients. A total of 299 CAP patients (including 97 severe CAP patients [SCAP]) and 20 healthy controls (HC) were included. A quantitative enzyme-linked immunosorbent test kit was employed for detecting the LPCAT level in plasma. We developed a deep-learning-based binary classification (SCAP or non-severe CAP [NSCAP]) model to process LPCAT levels and other laboratory test results. Results The level of LPCAT in patients with SCAP and death outcome was significantly higher than that in other patients. LPCAT showed the highest predictive value for SCAP. LPCAT was able to predict 30-day mortality among CAP patients, combining LPCAT values with PSI scores or CURB-65 further enhance mortality prediction accuracy. Conclusion The on admission level of LPCAT found significantly raised among SCAP patients and strongly predicted SCAP patients but with no correlation to etiology. Combining the LPCAT value with CURB-65 or PSI improved the 30-day mortality forecast significantly. Trial registration NCT03093220 Registered on March 28th, 2017.
Collapse
Affiliation(s)
- Li Chen
- Department of Respiratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Jianbo Xue
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Lili Zhao
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Yukun He
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Shining Fu
- Department of Respiratory and Critical Care Medicine, Beijing Jishuitan Hospital, The Fourth Medical College of Peking University, Beijing, China
| | - Xinqian Ma
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Wenyi Yu
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Yanfen Tang
- Department of Respiratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Department of Respiratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhancheng Gao
- Department of Respiratory & Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
6
|
Valentine WJ, Shimizu T, Shindou H. Lysophospholipid acyltransferases orchestrate the compositional diversity of phospholipids. Biochimie 2023; 215:24-33. [PMID: 37611890 DOI: 10.1016/j.biochi.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023]
Abstract
Lysophospholipid acyltransferases (LPLATs), in concert with glycerol-3-phosphate acyltransferases (GPATs) and phospholipase A1/2s, orchestrate the compositional diversity of the fatty chains in membrane phospholipids. Fourteen LPLAT enzymes which come from two distinct families, AGPAT and MBOAT, have been identified, and in this mini-review we provide an overview of their roles in de novo and remodeling pathways of membrane phospholipid biosynthesis. Recently new nomenclature for LPLATs has been introduced (LPLATx, where x is a number 1-14), and we also give an overview of key biological functions that have been discovered for LPLAT1-14, revealed primarily through studies of LPLAT-gene-deficient mice as well as by linkages to various human diseases.
Collapse
Affiliation(s)
- William J Valentine
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, 187-8502, Japan.
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo, 162-8655, Japan; Institute of Microbial Chemistry, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo, 162-8655, Japan; Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
7
|
He M, Li Z, Tung VSK, Pan M, Han X, Evgrafov O, Jiang XC. Inhibiting Phosphatidylcholine Remodeling in Adipose Tissue Increases Insulin Sensitivity. Diabetes 2023; 72:1547-1559. [PMID: 37625119 PMCID: PMC10588299 DOI: 10.2337/db23-0317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cell membrane phosphatidylcholine (PC) composition is regulated by lysophosphatidylcholine acyltransferase (LPCAT); changes in membrane PC saturation are implicated in metabolic disorders. Here, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue and created adipocyte-specific Lpcat3-knockout mice to study adipose tissue lipid metabolism. Transcriptome sequencing and plasma adipokine profiling were used to investigate how LPCAT3 regulates adipose tissue insulin signaling. LPCAT3 deficiency reduced polyunsaturated PCs in adipocyte plasma membranes, increasing insulin sensitivity. LPCAT3 deficiency influenced membrane lipid rafts, which activated insulin receptors and AKT in adipose tissue, and attenuated diet-induced insulin resistance. Conversely, higher LPCAT3 activity in adipose tissue from ob/ob, db/db, and high-fat diet-fed mice reduced insulin signaling. Adding polyunsaturated PCs to mature human or mouse adipocytes in vitro worsened insulin signaling. We suggest that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mulin He
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Zhiqiang Li
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Victoria Sook Keng Tung
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Meixia Pan
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xianlin Han
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Oleg Evgrafov
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Xian-Cheng Jiang
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
- Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, New York, NY
| |
Collapse
|
8
|
Guo PP, Jin X, Zhang JF, Li Q, Yan CG, Li XZ. Overexpression of DGAT2 Regulates the Differentiation of Bovine Preadipocytes. Animals (Basel) 2023; 13:ani13071195. [PMID: 37048451 PMCID: PMC10093762 DOI: 10.3390/ani13071195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
Triacylglycerols (TAGs) are a major component of intramuscular fat. Diacylglycerol O-acyltransferase 2(DGAT2) expression determines the rate of TAG synthesis. The purpose of this study was to investigate the role of DGAT2 in the differentiation of Yanbian cattle preadipocytes and lipid metabolism-related signalling pathways. Bovine preadipocytes were infected with overexpression and interfering adenovirus vectors of DGAT2. The effects on the differentiation of Yanbian cattle preadipocytes were examined using molecular and transcriptomic techniques, including differentially expressed genes (DEGs) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis. DGAT2 overexpression significantly increased (p < 0.05) intracellular TAG, adiponectin, and lipid droplet (LD) contents. Moreover, it upregulated (p < 0.05) peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer binding protein α, and fatty acid binding protein 4 mRNA expression. In contrast, DGAT2 knockdown reduced intracellular TAG and LD content and downregulated (p < 0.05) C/EBPβ, mannosyl (alpha-1,3-)-glycoproteinbeta-1,2-N-acetylglucosaminyltransferase, lipin 1,1-acylglycerol-3-phosphate O-acyltransferase 4, and acetyl-CoA carboxylase alpha mRNA expression. Between DGAT2-overexpressing preadipocytes and normal cells, 208 DEGs were identified, including 106 upregulated and 102 downregulated genes. KEGG pathway analysis revealed DEGs mainly enriched in PPAR signalling and AMP-activated protein kinase pathways, cholesterol metabolism, and fatty acid biosynthesis. These results demonstrated that DGAT2 regulated preadipocyte differentiation and LD and TAG accumulation by mediating the expression of adipose differentiation-, lipid metabolism-, and fatty acid synthesis-related genes.
Collapse
|
9
|
Ding Y, Cui K, Han S, Hao T, Liu Y, Lai W, Xu X, Mai K, Ai Q. Lysophosphatidylcholine acyltransferase 3 (LPCAT3) mediates palmitate-induced inflammation in macrophages of large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2022; 126:12-20. [PMID: 35526799 DOI: 10.1016/j.fsi.2022.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
LPCAT3, a subtype of lysophosphatidylcholine acyltransferases, is a key enzyme in phosphatidylcholine remodeling pathway and plays a significant role in mediating inflammatory response in mammals. However, its inflammatory function in fish has yet to be discovered. Herein, this study aimed to investigate its role in inflammation in Larimichthys crocea. We analyzed the coding sequence of Larimichthys crocea LPCAT3 (Lc-LPCAT3) and explored the effect of Lc-LPCAT3 on palmitate (PA)-induced inflammation. We found that in macrophage cell line of Larimichthys crocea, the mRNA expression of Lc-lpcat3 was upregulated by PA with the elevated pro-inflammatory genes expression, including il1β, il6, il8, tnfα and ifnγ. Next, the role of Lc-LPCAT3 in inflammation induced by PA was further investigated. Results showed that knockdown of Lc-LPCAT3 mitigated PA-induced pro-inflammatory genes mRNA expression, including il1β, il8, tnfα and ifnγ, in which JNK signaling pathway was involved. In contrast, overexpression of Lc-LPCAT3 induced pro-inflammatory genes expression including il1β, tnfα and ifnγ. Furthermore, several transcription factors with negative regulation of Lc-LPCAT3 promoter activity were discovered including LXRα, RXRα, PPARα, PPARγ, CEBPα, CEBPβ, CEBPδ, SREBP1 and SREBP2, and SREBP1 had the strongest regulatory effect. In conclusion, we first discovered that fish LPCAT3 participated in PA-induced inflammation, and targeting SREBP1 might be an effective coping strategy.
Collapse
Affiliation(s)
- Yi Ding
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Kun Cui
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Shangzhe Han
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Tingting Hao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Yongtao Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Wencong Lai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Xiang Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266003, Qingdao, Shandong, PR China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affair), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266003, Qingdao, Shandong, PR China.
| |
Collapse
|
10
|
Zhang Z, Zheng R, Zhu C, Geng H, Xu G. Lipidomics characterization of the lipid metabolism profiles in a cystinuria rat model: Precalculus damage in the kidney of cystinuria. Prostaglandins Other Lipid Mediat 2022; 162:106651. [PMID: 35680078 DOI: 10.1016/j.prostaglandins.2022.106651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/20/2022] [Accepted: 06/02/2022] [Indexed: 10/18/2022]
Abstract
Cystinuria is a genetic disorder of cystine transport, including defective protein b0,+AT (encoded by SLC7A9), and/or rBAT (encoded by SLC3A1). Patients present hyperexcretion of cystine in the urine, recurrent cystine lithiasis, and progressive decline in kidney function. Moreover, heterodimer transport is defective. To date, little omics data are accessible regarding this metabolic disease caused by membrane proteins. Since membrane function is closely related to changes in the lipidome, we decided to explore the changes in kidney tissue of a self-established cystinuria rat model by performing lipidomic analysis by LC-MS/MS. Our results demonstrated that Slc7a9 deficiency changed the lipid profile of the renal cortex and induced vital modifications in the lipidome, including major alterations in ChE, LPA, and PA. Among those alterations, this lipidomic study highlights the lipid changes that participate in inflammatory responses during cystinuria. As a result, lipid research, perhaps has great potential, for it may lead to the identification of novel therapeutic targets for the prevention and treatment of cystinuria.
Collapse
Affiliation(s)
- Zihan Zhang
- Shanghai Jiaotong University School of Medicine, China
| | - Rui Zheng
- Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Caihua Zhu
- Shanghai Applied Protein Technology Co., Ltd., 201100, China
| | - Hongquan Geng
- Shanghai Jiaotong University School of Medicine, China; Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China.
| | - Guofeng Xu
- Shanghai Jiaotong University School of Medicine, China; Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China.
| |
Collapse
|
11
|
Tang CH, Shi SH, Lin CY, Wang WH. Lipid profiling differentiates the effect of ambient microenriched copper on a coral as an advanced tool for biomonitoring. MARINE POLLUTION BULLETIN 2022; 178:113650. [PMID: 35447438 DOI: 10.1016/j.marpolbul.2022.113650] [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: 12/01/2021] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
Copper can be beneficial or harmful to coral at environmentally relevant levels, making environmental monitoring a challenging. Membrane lipids make the cell a dynamic environment according to the circumstances; thus, the lipid profile should be indicative of an environmental/physiological state. To gain more insight into the copper effect on coral health and be a basis of biomonitoring, glycerophosphocholine profiling of coral exposed to microenriched copper levels was conducted in this study. The copper microenrichments resulted in a diacritical effect of decreasing carbonic anhydrase activity, following a supplementation effect, on coral lipid metabolism. Microdifferences in copper levels are critical to determine the coral metabolic state and were therefore included in this study. In addition, an excellent quantitative model correlating the coral lipid variation with the exposed copper levels or the induced physiological effect was obtained to demonstrate its performance for biomonitoring.
Collapse
Affiliation(s)
- Chuan-Ho Tang
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Shu-Han Shi
- Institute of Marine Biology, National Dong Hwa University, Pingtung, Taiwan
| | - Ching-Yu Lin
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taiwan
| | - Wei-Hsien Wang
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
12
|
Shao G, Qian Y, Lu L, Liu Y, Wu T, Ji G, Xu H. Research progress in the role and mechanism of LPCAT3 in metabolic related diseases and cancer. J Cancer 2022; 13:2430-2439. [PMID: 35711841 PMCID: PMC9174858 DOI: 10.7150/jca.71619] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Lysophosphatidylcholine acyltransferases (LPCATs) are among the lysophopholipid acyltransferases (LPLATs) that specifically regulate the abundance of different phosphatidylcholine (PC) species in a variety of cell and tissue types, thereby playing an important role in lipid metabolism and homeostasis. Lysophosphatidylcholine acyltransferase 3 (LPCAT3, MBOAT5) is a member of the LPCAT family that primarily regulates the levels of arachidonic PC species. LPCAT3 is regulated by the liver X receptor, which plays an important role in lipoprotein production in the liver and small intestine. Increasing lines of research have demonstrated that LPCAT3 plays important roles in the occurrence and development of many diseases, such as atherosclerosis, intestinal tumors, and nonalcoholic steatohepatitis (NASH). The development of many diseases has been linked to the proinflammatory effects of LPCAT3. This review focuses on the current knowledge of LPCAT3, including its function and mechanism in different diseases. We aim to provide a comprehensive and in-depth understanding of LPCAT3 and to provide new ideas for the treatment of some diseases.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| | - Yufan Qian
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine - Shanghai 200032, China
| |
Collapse
|
13
|
Valentine WJ, Yanagida K, Kawana H, Kono N, Noda NN, Aoki J, Shindou H. Update and nomenclature proposal for mammalian lysophospholipid acyltransferases which create membrane phospholipid diversity. J Biol Chem 2021; 298:101470. [PMID: 34890643 PMCID: PMC8753187 DOI: 10.1016/j.jbc.2021.101470] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
The diversity of glycerophospholipid species in cellular membranes is immense and affects various biological functions. Glycerol-3-phosphate acyltransferases (GPATs) and lysophospholipid acyltransferases (LPLATs), in concert with phospholipase A1/2s enzymes, contribute to this diversity via selective esterification of fatty acyl chains at the sn-1 or sn-2 positions of membrane phospholipids. These enzymes are conserved across all kingdoms, and in mammals four GPATs of the 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) family and at least 14 LPLATs, either of the AGPAT or the membrane-bound O-acyltransferase (MBOAT) families, have been identified. Here we provide an overview of the biochemical and biological activities of these mammalian enzymes, including their predicted structures, involvements in human diseases, and essential physiological roles as revealed by gene-deficient mice. Recently, the nomenclature used to refer to these enzymes has generated some confusion due to the use of multiple names to refer to the same enzyme and instances of the same name being used to refer to completely different enzymes. Thus, this review proposes a more uniform LPLAT enzyme nomenclature, as well as providing an update of recent advances made in the study of LPLATs, continuing from our JBC mini review in 2009.
Collapse
Affiliation(s)
- William J Valentine
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo 162-8655, Japan; Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, 187-8502, Japan
| | - Keisuke Yanagida
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nobuo N Noda
- Institute of Microbial Chemistry (BIKAKEN), Microbial Chemistry Research Foundation, Tokyo 141-0021, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo 162-8655, Japan; Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
14
|
Xiang H, Shao M, Lu Y, Wang J, Wu T, Ji G. Kaempferol Alleviates Steatosis and Inflammation During Early Non-Alcoholic Steatohepatitis Associated With Liver X Receptor α-Lysophosphatidylcholine Acyltransferase 3 Signaling Pathway. Front Pharmacol 2021; 12:690736. [PMID: 34262459 PMCID: PMC8273916 DOI: 10.3389/fphar.2021.690736] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Kaempferol (KP) has a variety of biological effects such as anti-inflammatory, anti-oxidant, anti-aging and cardiovascular protection. Whether KP has a therapeutic effect on non-alcoholic steatohepatitis (NASH), and the detailed mechanism is currently unclear. This study aims to explore the mechanism of KP in the treatment of NASH through in vivo and in vitro experiments. Methods: 1) In vivo experiment: In the C57BL/6 NASH mice model induced by high fat diet (HFD), KP was administered by gavage at a dose of 20 mg/kg/day. 2) In vitro experiment: Palmitic acid/Oleic acid (PA/OA, 0.375/0.75 mM) was used to intervene HepG2 and AML12 cells to establish a steatosis cell model. Three concentrations of KP, low (20 μmol/L), medium (40 μmol/L) and high (60 μmol/L) were used in vitro. The mRNA and protein expression of related molecules involved in LXRα-LPCAT3-ERS pathway were detected using RT-qPCR and Western blot. Results: In the NASH mouse model, KP can significantly reduce the expression of LXRα, LPCAT3 and ERS-related factors PERK, eIF2α, ATF6, ATF4, XBP1, CHOP, IRE1α and GRP78. In the PA/OA-induced cell model, KP could decrease the content of triglyceride and lipid droplets, and also decrease the expression of LXR α, LPCAT3 and ERS related factors PERK, eIF2α, ATF6, ATF4, XBP1, CHOP, IRE1α and GRP78. Conclusion: KP may decrease the expression level of LXRα and LPCAT3, thus improve ERS and reduce hepatic steatosis and inflammation.
Collapse
Affiliation(s)
- Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Shao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Immunohistochemical staining reveals differential expression of ACSL3 and ACSL4 in hepatocellular carcinoma and hepatic gastrointestinal metastases. Biosci Rep 2021; 40:222647. [PMID: 32286604 PMCID: PMC7198044 DOI: 10.1042/bsr20200219] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Long-chain fatty acyl CoA synthetases (ACSLs) activate fatty acids by CoA addition thus facilitating their intracellular metabolism. Dysregulated ACSL expression features in several cancers and can affect processes such as ferroptosis, fatty acid β-oxidation, prostaglandin biosynthesis, steroidogenesis and phospholipid acyl chain remodelling. Here we investigate long chain acyl-CoA synthetase 3 (ACSL3) and long chain acyl-CoA synthetase 4 (ACSL4) expression in liver malignancies. The expression and subcellular localisations of the ACSL3 and ACSL4 isoforms in hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA) and hepatic metastases were assessed by immunohistochemical analyses of multiple tumour tissue arrays and by subcellular fractionation of cultured HepG2 cells. The expression of both enzymes was increased in HCC compared with normal liver. Expression of ACSL3 was similar in HCC and hepatic metastases but lower in healthy tissue. Increased ACSL3 expression distinguished HCC from CCA with a sensitivity of 87.2% and a specificity of 75%. ACSL4 expression was significantly greater in HCC than in all other tumours and distinguished HCC from normal liver tissue with a sensitivity of 93.8% and specificity of 93.6%. Combined ACSL3 and ACSL4 staining scores distinguished HCC from hepatic metastases with 80.1% sensitivity and 77.1% specificity. These enzymes had partially overlapping intracellular distributions, ACSL4 localised to the plasma membrane and both isoforms associated with lipid droplets and the endoplasmic reticulum (ER). In conclusion, analysis of ACSL3 and ACSL4 expression can distinguish different classes of hepatic tumours.
Collapse
|
16
|
Lewandowski CT, Khan MW, BenAissa M, Dubrovskyi O, Ackerman-Berrier M, LaDu MJ, Layden BT, Thatcher GRJ. Metabolomic analysis of a selective ABCA1 inducer in obesogenic challenge provides a rationale for therapeutic development. EBioMedicine 2021; 66:103287. [PMID: 33752129 PMCID: PMC8010624 DOI: 10.1016/j.ebiom.2021.103287] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Therapeutic agents with novel mechanisms of action are needed to combat the growing epidemic of type 2 diabetes (T2D) and related metabolic syndromes. Liver X receptor (LXR) agonists possess preclinical efficacy yet produce side effects due to excessive lipogenesis. Anticipating that many beneficial and detrimental effects of LXR agonists are mediated by ABCA1 and SREPB1c expression, respectively, we hypothesized that a phenotypic optimization strategy prioritizing selective ABCA1 induction would identify an efficacious lead compound with an improved side effect profile over existing LXRβ agonists. METHODS We synthesized and characterized a novel small molecule for selective induction of ABCA1 vs. SREBP1c in vitro. This compound was evaluated in both wild-type mice and a high-fat diet (HFD) mouse model of obesity-driven diabetes through functional, biochemical, and metabolomic analysis. FINDINGS Six weeks of oral administration of our lead compound attenuated weight gain, glucose intolerance, insulin signaling deficits, and adiposity. Global metabolomics revealed suppression of gluconeogenesis, free fatty acids, and pro-inflammatory metabolites. Target identification linked these beneficial effects to selective LXRβ agonism and PPAR/RXR antagonism. INTERPRETATION Our observations in the HFD model, combined with the absence of lipogenesis and neutropenia in WT mice, support this novel approach to therapeutic development for T2D and related conditions.
Collapse
Affiliation(s)
- Cutler T Lewandowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Md Wasim Khan
- Department of Medicine, University of Illinois at Chicago, 835 S. Wolcott St., Chicago, IL 60612, USA
| | - Manel BenAissa
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Oleksii Dubrovskyi
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Martha Ackerman-Berrier
- Department of Pharmacology and Toxicology, University of Arizona, 1295N. Martin, Tucson, AZ 85721, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Brian T Layden
- Department of Medicine, University of Illinois at Chicago, 835 S. Wolcott St., Chicago, IL 60612, USA.
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, University of Arizona, 1295N. Martin, Tucson, AZ 85721, USA.
| |
Collapse
|
17
|
Armitage EG, Barnes A, Patrick K, Bechar J, Harrison MJ, Lavery GG, Rainger GE, Buckley CD, Loftus NJ, Wilson ID, Naylor AJ. Metabolic consequences for mice lacking Endosialin: LC-MS/MS-based metabolic phenotyping of serum from C56Bl/6J Control and CD248 knock-out mice. Metabolomics 2021; 17:14. [PMID: 33462674 PMCID: PMC7813710 DOI: 10.1007/s11306-020-01764-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/22/2020] [Indexed: 11/17/2022]
Abstract
INTRODUCTION The Endosialin/CD248/TEM1 protein is expressed in adipose tissue and its expression increases with obesity. Recently, genetic deletion of CD248 has been shown to protect mice against atherosclerosis on a high fat diet. OBJECTIVES We investigated the effect of high fat diet feeding on visceral fat pads and circulating lipid profiles in CD248 knockout mice compared to controls. METHODS From 10 weeks old, CD248-/- and +/+ mice were fed either chow (normal) diet or a high fat diet for 13 weeks. After 13 weeks the metabolic profiles and relative quantities of circulating lipid species were assessed using ultra high performance liquid chromatography-quadrupole time-of flight mass spectrometry (UHPLC-MS) with high resolution accurate mass (HRAM) capability. RESULTS We demonstrate a specific reduction in the size of the perirenal fat pad in CD248-/- mice compared to CD248+/+, despite similar food intake. More strikingly, we identify significant, diet-dependent differences in the serum metabolic phenotypes of CD248 null compared to age and sex-matched wildtype control mice. Generalised protection from HFD-induced lipid accumulation was observed in CD248 null mice compared to wildtype, with particular reduction noted in the lysophosphatidylcholines, phosphatidylcholines, cholesterol and carnitine. CONCLUSIONS Overall these results show a clear and protective metabolic consequence of CD248 deletion in mice, implicating CD248 in lipid metabolism or trafficking and opening new avenues for further investigation using anti-CD248 targeting agents.
Collapse
Affiliation(s)
| | | | - Kieran Patrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Janak Bechar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Matthew J Harrison
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - G Ed Rainger
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Christopher D Buckley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, UK
| | | | - Ian D Wilson
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, UK
| | - Amy J Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
| |
Collapse
|
18
|
Bourgeois T, Jalil A, Thomas C, Magnani C, Le Guern N, Gautier T, Pais de Barros JP, Bergas V, Choubley H, Mazzeo L, Menegaut L, Josiane Lebrun L, Van Dongen K, Xolin M, Jourdan T, Buch C, Labbé J, Saas P, Lagrost L, Masson D, Grober J. Deletion of lysophosphatidylcholine acyltransferase 3 in myeloid cells worsens hepatic steatosis after a high-fat diet. J Lipid Res 2020; 62:100013. [PMID: 33518513 PMCID: PMC7859853 DOI: 10.1194/jlr.ra120000737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/25/2020] [Accepted: 12/11/2020] [Indexed: 12/28/2022] Open
Abstract
Recent studies have highlighted an important role for lysophosphatidylcholine acyltransferase 3 (LPCAT3) in controlling the PUFA composition of cell membranes in the liver and intestine. In these organs, LPCAT3 critically supports cell-membrane-associated processes such as lipid absorption or lipoprotein secretion. However, the role of LPCAT3 in macrophages remains controversial. Here, we investigated LPCAT3's role in macrophages both in vitro and in vivo in mice with atherosclerosis and obesity. To accomplish this, we used the LysMCre strategy to develop a mouse model with conditional Lpcat3 deficiency in myeloid cells (Lpcat3KOMac). We observed that partial Lpcat3 deficiency (approximately 75% reduction) in macrophages alters the PUFA composition of all phospholipid (PL) subclasses, including phosphatidylinositols and phosphatidylserines. A reduced incorporation of C20 PUFAs (mainly arachidonic acid [AA]) into PLs was associated with a redistribution of these FAs toward other cellular lipids such as cholesteryl esters. Lpcat3 deficiency had no obvious impact on macrophage inflammatory response or endoplasmic reticulum (ER) stress; however, Lpcat3KOMac macrophages exhibited a reduction in cholesterol efflux in vitro. In vivo, myeloid Lpcat3 deficiency did not affect atherosclerosis development in LDL receptor deficient mouse (Ldlr-/-) mice. Lpcat3KOMac mice on a high-fat diet displayed a mild increase in hepatic steatosis associated with alterations in several liver metabolic pathways and in liver eicosanoid composition. We conclude that alterations in AA metabolism along with myeloid Lpcat3 deficiency may secondarily affect AA homeostasis in the whole liver, leading to metabolic disorders and triglyceride accumulation.
Collapse
Affiliation(s)
- Thibaut Bourgeois
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Antoine Jalil
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Charlène Magnani
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Naig Le Guern
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Thomas Gautier
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Jean-Paul Pais de Barros
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; Lipidomic analytic plate-forme, Univ. Bourgogne Franche-Comté, Batiment B3, Bvd Maréchal de Lattre de Tassigny, Dijon, France
| | - Victoria Bergas
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; Lipidomic analytic plate-forme, Univ. Bourgogne Franche-Comté, Batiment B3, Bvd Maréchal de Lattre de Tassigny, Dijon, France
| | - Hélène Choubley
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; Lipidomic analytic plate-forme, Univ. Bourgogne Franche-Comté, Batiment B3, Bvd Maréchal de Lattre de Tassigny, Dijon, France
| | - Loïc Mazzeo
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Louise Menegaut
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Lorène Josiane Lebrun
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; AgroSup Dijon, Dijon, France
| | - Kévin Van Dongen
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Marion Xolin
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Tony Jourdan
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Chloé Buch
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Jérome Labbé
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Philippe Saas
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte Greffon-Tumeur/Ingénierie Cellulaire et Génique, LabEx LipSTIC, Besançon, France
| | - Laurent Lagrost
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; CHU Dijon, laboratoire de Biochimie, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; CHU Dijon, laboratoire de Biochimie, Dijon, France
| | - Jacques Grober
- Univ. Bourgogne Franche-Comté, LNC UMR12131, Dijon, France; INSERM, LNC UMR 1231, Dijon, France; FCS Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France; AgroSup Dijon, Dijon, France.
| |
Collapse
|
19
|
Abstract
The cause of insulin resistance in obesity and type 2 diabetes mellitus (T2DM) is not limited to impaired insulin signalling but also involves the complex interplay of multiple metabolic pathways. The analysis of large data sets generated by metabolomics and lipidomics has shed new light on the roles of metabolites such as lipids, amino acids and bile acids in modulating insulin sensitivity. Metabolites can regulate insulin sensitivity directly by modulating components of the insulin signalling pathway, such as insulin receptor substrates (IRSs) and AKT, and indirectly by altering the flux of substrates through multiple metabolic pathways, including lipogenesis, lipid oxidation, protein synthesis and degradation and hepatic gluconeogenesis. Moreover, the post-translational modification of proteins by metabolites and lipids, including acetylation and palmitoylation, can alter protein function. Furthermore, the role of the microbiota in regulating substrate metabolism and insulin sensitivity is unfolding. In this Review, we discuss the emerging roles of metabolites in the pathogenesis of insulin resistance and T2DM. A comprehensive understanding of the metabolic adaptations involved in insulin resistance may enable the identification of novel targets for improving insulin sensitivity and preventing, and treating, T2DM.
Collapse
|
20
|
Du X, Hu J, Zhang Q, Liu Q, Xiang X, Dong J, Lou B, He S, Gu X, Cao Y, Li Y, Ding T. A novel assay for measuring recombinant human lysophosphatidylcholine acyltransferase 3 activity. FEBS Open Bio 2019; 9:1734-1743. [PMID: 31376210 PMCID: PMC6768109 DOI: 10.1002/2211-5463.12712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/02/2019] [Accepted: 08/02/2019] [Indexed: 11/28/2022] Open
Abstract
Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is an important enzyme in phospholipid remodeling, a process that influences the biophysical properties of cell membranes and thus cell function. Multiple lines of evidence suggest that LPCAT3 is involved in several diseases, including atherosclerosis, non‐alcoholic steatohepatitis, and carcinoma. Thus, LPCAT3 may have potential as a therapeutic target for these diseases. In the present study, we devised an assay based on reversed‐phase HPLC to measure LPCAT3 activity, which may facilitate the identification of LPCAT3 inhibitors and activators. We found that optimal pH and temperature of recombinant human LPCAT3 are 6.0 and 30 °C, respectively. The enzyme Km values for substrates NBD‐labelled lysophosphatidylcholine and arachidonoyl CoA were 266.84 ± 3.65 and 11.03 ± 0.51 μmol·L−1, respectively, and the Vmax was 39.76 ± 1.86 pmol·min−1·U−1. Moreover, we used our new method to determine the IC50 of a known LPCAT inhibitor, TSI‐10. In conclusion, this novel assay can be used to measure the effects of compounds on LPCAT3 activity.
Collapse
Affiliation(s)
- Xinming Du
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiachun Hu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Qing Zhang
- Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qi Liu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Xinxin Xiang
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Jibin Dong
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Bin Lou
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Shuhua He
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiang Gu
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Yu Cao
- Institute of Precision Medicine, The Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yingxia Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Tingbo Ding
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Singh AB, Kan CFK, Kraemer FB, Sobel RA, Liu J. Liver-specific knockdown of long-chain acyl-CoA synthetase 4 reveals its key role in VLDL-TG metabolism and phospholipid synthesis in mice fed a high-fat diet. Am J Physiol Endocrinol Metab 2019; 316:E880-E894. [PMID: 30721098 PMCID: PMC6580179 DOI: 10.1152/ajpendo.00503.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long-chain acyl-CoA synthetase 4 (ACSL4) has a unique substrate specificity for arachidonic acid. Hepatic ACSL4 is coregulated with the phospholipid (PL)-remodeling enzyme lysophosphatidylcholine (LPC) acyltransferase 3 by peroxisome proliferator-activated receptor δ to modulate the plasma triglyceride (TG) metabolism. In this study, we investigated the acute effects of hepatic ACSL4 deficiency on lipid metabolism in adult mice fed a high-fat diet (HFD). Adenovirus-mediated expression of a mouse ACSL4 shRNA (Ad-shAcsl4) in the liver of HFD-fed mice led to a 43% reduction of hepatic arachidonoyl-CoA synthetase activity and a 53% decrease in ACSL4 protein levels compared with mice receiving control adenovirus (Ad-shLacZ). Attenuated ACSL4 expression resulted in a substantial decrease in circulating VLDL-TG levels without affecting plasma cholesterol. Lipidomics profiling revealed that knocking down ACSL4 altered liver PL compositions, with the greatest impact on accumulation of abundant LPC species (LPC 16:0 and LPC 18:0) and lysophosphatidylethanolamine (LPE) species (LPE 16:0 and LPE 18:0). In addition, fasting glucose and insulin levels were higher in Ad-shAcsl4-transduced mice versus control (Ad-shLacZ). Glucose tolerance testing further indicated an insulin-resistant phenotype upon knockdown of ACSL4. These results provide the first in vivo evidence that ACSL4 plays a role in plasma TG and glucose metabolism and hepatic PL synthesis of hyperlipidemic mice.
Collapse
Affiliation(s)
- Amar B Singh
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Chin Fung K Kan
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Ochsner Clinical School, University of Queensland School of Medicine , New Orleans, Louisiana
| | - Fredric B Kraemer
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Department of Medicine, Stanford University School of Medicine , Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine , Stanford, California
| | - Raymond A Sobel
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Department of Pathology, Stanford University School of Medicine , Stanford, California
| | - Jingwen Liu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
22
|
Law SH, Chan ML, Marathe GK, Parveen F, Chen CH, Ke LY. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. Int J Mol Sci 2019; 20:ijms20051149. [PMID: 30845751 PMCID: PMC6429061 DOI: 10.3390/ijms20051149] [Citation(s) in RCA: 499] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidylcholine (LPC) is increasingly recognized as a key marker/factor positively associated with cardiovascular and neurodegenerative diseases. However, findings from recent clinical lipidomic studies of LPC have been controversial. A key issue is the complexity of the enzymatic cascade involved in LPC metabolism. Here, we address the coordination of these enzymes and the derangement that may disrupt LPC homeostasis, leading to metabolic disorders. LPC is mainly derived from the turnover of phosphatidylcholine (PC) in the circulation by phospholipase A2 (PLA2). In the presence of Acyl-CoA, lysophosphatidylcholine acyltransferase (LPCAT) converts LPC to PC, which rapidly gets recycled by the Lands cycle. However, overexpression or enhanced activity of PLA2 increases the LPC content in modified low-density lipoprotein (LDL) and oxidized LDL, which play significant roles in the development of atherosclerotic plaques and endothelial dysfunction. The intracellular enzyme LPCAT cannot directly remove LPC from circulation. Hydrolysis of LPC by autotaxin, an enzyme with lysophospholipase D activity, generates lysophosphatidic acid, which is highly associated with cancers. Although enzymes with lysophospholipase A1 activity could theoretically degrade LPC into harmless metabolites, they have not been found in the circulation. In conclusion, understanding enzyme kinetics and LPC metabolism may help identify novel therapeutic targets in LPC-associated diseases.
Collapse
Affiliation(s)
- Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Mei-Lin Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, MacKay Medical College, Taipei 10449, Taiwan.
| | - Gopal K Marathe
- Department of Studies in Biochemistry, Manasagangothri, University of Mysore, Mysore-570006, India.
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA.
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
23
|
Singh AB, Dong B, Xu Y, Zhang Y, Liu J. Identification of a novel function of hepatic long-chain acyl-CoA synthetase-1 (ACSL1) in bile acid synthesis and its regulation by bile acid-activated farnesoid X receptor. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:358-371. [PMID: 30580099 DOI: 10.1016/j.bbalip.2018.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022]
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) plays a pivotal role in fatty acid β‑oxidation in heart, adipose tissue and skeletal muscle. However, key functions of ACSL1 in the liver remain largely unknown. We investigated acute effects of hepatic ACSL1 deficiency on lipid metabolism in adult mice under hyperlipidemic and normolipidemic conditions. We knocked down hepatic ACSL1 expression using adenovirus expressing a ACSL1 shRNA (Ad-shAcsl1) in mice fed a high-fat diet or a normal chow diet. Hepatic ACSL1 depletion generated a hypercholesterolemic phenotype in mice fed both diets with marked elevations of total cholesterol, LDL-cholesterol and free cholesterol in circulation and accumulations of cholesterol in the liver. Furthermore, SREBP2 pathway in ACSL1 depleted livers was severely repressed with a 50% reduction of LDL receptor protein levels. In contrast to the dysregulated cholesterol metabolism, serum triglycerides, free fatty acid and phospholipid levels were unaffected. Mechanistic investigations of genome-wide gene expression profiling and pathway analysis revealed that ACSL1 depletion repressed expressions of several key enzymes for bile acid biosynthesis, consequently leading to reduced liver bile acid levels and altered bile acid compositions. These results are the first demonstration of a requisite role of ACSL1 in bile acid biosynthetic pathway in liver tissue. Furthermore, we discovered that Acsl1 is a novel molecular target of the bile acid-activated farnesoid X receptor (FXR). Activation of FXR by agonist obeticholic acid repressed the expression of ACSL1 protein and mRNA in the liver of FXR wild-type mice but not in FXR knockout mice.
Collapse
Affiliation(s)
- Amar Bahadur Singh
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America
| | - Bin Dong
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America
| | - Yanyong Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Jingwen Liu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, United States of America.
| |
Collapse
|
24
|
Abstract
Phospholipids are major constituents of biological membranes. The fatty acyl chain composition of phospholipids determines the biophysical properties of membranes and thereby affects their impact on biological processes. The composition of fatty acyl chains is also actively regulated through a deacylation and reacylation pathway called Lands' cycle. Recent studies of mouse genetic models have demonstrated that lysophosphatidylcholine acyltransferases (LPCATs), which catalyze the incorporation of fatty acyl chains into the sn-2 site of phosphatidylcholine, play important roles in pathophysiology. Two LPCAT family members, LPCAT1 and LPCAT3, have been particularly well studied. LPCAT1 is crucial for proper lung function due to its role in pulmonary surfactant biosynthesis. LPCAT3 maintains systemic lipid homeostasis by regulating lipid absorption in intestine, lipoprotein secretion, and de novo lipogenesis in liver. Mounting evidence also suggests that changes in LPCAT activity may be potentially involved in pathological conditions, including nonalcoholic fatty liver disease, atherosclerosis, viral infections, and cancer. Pharmacological manipulation of LPCAT activity and membrane phospholipid composition may provide new therapeutic options for these conditions.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90272, USA;
| |
Collapse
|
25
|
Thomas C, Jalil A, Magnani C, Ishibashi M, Queré R, Bourgeois T, Bergas V, Ménégaut L, Patoli D, Le Guern N, Labbé J, Gautier T, de Barros JPP, Lagrost L, Masson D. LPCAT3 deficiency in hematopoietic cells alters cholesterol and phospholipid homeostasis and promotes atherosclerosis. Atherosclerosis 2018; 275:409-418. [PMID: 29866392 DOI: 10.1016/j.atherosclerosis.2018.05.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/04/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS LPCAT3 plays a major role in phospholipid metabolism in the liver and intestine. However, the impact of LPCAT3 on hematopoietic cell and macrophage functions has yet to be described. Our aim was to understand the functions of LPCAT3 in macrophages and to investigate whether LPCAT3 deficiency in hematopoietic cells may affect atherosclerosis development. METHODS Mice with constitutive Lpcat3 deficiency (Lpcat3-/-) were generated. We used fetal hematopoietic liver cells to generate WT and Lpcat3-/- macrophages in vitro and to perform hematopoietic cell transplantation in recipient Ldlr-/- mice. RESULTS Lpcat3-deficient macrophages displayed major reductions in the arachidonate content of phosphatidylcholines, phosphatidylethanolamines and, unexpectedly, plasmalogens. These changes were associated with altered cholesterol homeostasis, including an increase in the ratio of free to esterified cholesterol and a reduction in cholesterol efflux in Lpcat3-/- macrophages. This correlated with the inhibition of some LXR-regulated pathways, related to altered cellular availability of the arachidonic acid. Indeed, LPCAT3 deficiency was associated with decreased Abca1, Abcg1 and ApoE mRNA levels in fetal liver cells derived macrophages. In vivo, these changes translated into a significant increase in atherosclerotic lesions in Ldlr-/- mice with hematopoietic LPCAT3 deficiency. CONCLUSIONS This study identifies LPCAT3 as a key factor in the control of phospholipid homeostasis and arachidonate availability in myeloid cells and underlines a new role for LPCAT3 in plasmalogen metabolism. Moreover, our work strengthens the link between phospholipid and sterol metabolism in hematopoietic cells, with significant consequences on nuclear receptor-regulated pathways and atherosclerosis development.
Collapse
Affiliation(s)
- Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Antoine Jalil
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Charlène Magnani
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Minako Ishibashi
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Ronan Queré
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Thibaut Bourgeois
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Victoria Bergas
- Lipidomic analytic plate-forme, UBFC, Batiment B3, Bvd Maréchal de Lattre de Tassigny, 21000, Dijon, France
| | - Louise Ménégaut
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France; CHU Dijon, laboratoire de Biochimie, F-21000, Dijon, France
| | - Danish Patoli
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Naig Le Guern
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Jérôme Labbé
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Thomas Gautier
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Jean Paul Pais de Barros
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France; Lipidomic analytic plate-forme, UBFC, Batiment B3, Bvd Maréchal de Lattre de Tassigny, 21000, Dijon, France
| | - Laurent Lagrost
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France; CHU Dijon, laboratoire de Biochimie, F-21000, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; INSERM, LNC UMR 1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France; CHU Dijon, laboratoire de Biochimie, F-21000, Dijon, France.
| |
Collapse
|