1
|
Li H, Han Y, Chen W, Wang Y, Xu Y, Wang T, Gong J, Li W, Zhang H, Wang J, Qiu X, Zhu T. Lysoglycerophospholipid metabolism alterations associated with ambient fine particulate matter exposure: Insights into the pro-atherosclerotic effects. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125646. [PMID: 39761719 DOI: 10.1016/j.envpol.2025.125646] [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: 08/13/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
The biological pathways connecting ambient fine particulate matter (PM2.5)-induced initial adverse effects to the development of atherosclerotic cardiovascular diseases are not fully understood. We hypothesize that lysoglycerophospholipids (LysoGPLs) are pivotal mediators of atherosclerosis induced by exposure to PM2.5. This study investigated the changes of LysoGPLs in response to PM2.5 exposure and the mediation role of LysoGPLs in the pro-atherosclerotic effects of PM2.5 exposure. In this longitudinal panel study, 110 adults aged 50-65 years from Beijing, China, were followed between 2013 and 2015. Targeted metabolomics analyses were utilized to quantify 18 LysoGPLs from five subclasses in 579 plasma samples. Daily PM2.5 mass concentration was monitored at a station. We used linear mixed-effect models to estimate the responses of LysoGPLs to PM2.5 exposure. Subsequently, mediation analyses were conducted to investigate the mediating role of LysoGPLs in PM2.5-associated changes in non-high density lipoprotein-cholesterol (Non-HDL-C), a biomarker for pro-atherosclerotic apolipoprotein B-containing lipoproteins, and various inflammatory biomarkers, including interleukin (IL)-8, monocyte chemoattractant protein-1 (MCP-1), soluble CD40 ligand, and interferon (IFN)-γ. Short-to medium-term (1-30 days) PM2.5 exposure was associated with significant increases in six lysophosphatidic acids (LPAs), three lysoalkylphosphatidylcholines [LPC(O)s], and three lysophosphatidylglycerols (LPGs), as well as decreases in two LPAs and one lysophosphatidylserine (LysoPS), with maximus changes of 0.5-2.1%, 0.8-2.1%, 1.9-3.0%, -1.4-3.7%, and -8.0%, respectively. Furthermore, the elevated levels of LPA 18:1/18:2, LPC(O) 18:0/18:1, and LPG 16:0/16:1/18:0 significantly mediated the PM2.5-associated increase in Non-HDL-C (18-49%), IL-8 (9-24%), MCP-1 (12-26%), and IFN-γ (4-12%) over 30 days. In conclusion, short-to medium-term PM2.5 exposure was associated with altered metabolism of LysoGPLs, which mediated the PM2.5-associated pro-atherosclerotic response.
Collapse
Affiliation(s)
- Haonan Li
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China
| | - Yiqun Han
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China; Environmental Research Group, MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Wu Chen
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China; Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Yanwen Wang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China; National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yifan Xu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China
| | - Teng Wang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China; School of Health Policy and Management, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jicheng Gong
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China
| | - Weiju Li
- Peking University Hospital, Peking University, Beijing, China
| | - Hongyin Zhang
- Peking University Hospital, Peking University, Beijing, China
| | - Junxia Wang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China
| | - Xinghua Qiu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China
| | - Tong Zhu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing, China.
| |
Collapse
|
2
|
Sedghi M, Javanmard F, Amoozmehr A, Zamany S, Mohammadi I, Kim W, Choppa VSR. Lysophospholipid Supplementation in Broiler Breeders' Diet Benefits Offspring's Productive Performance, Blood Parameters, and Hepatic β-Oxidation Genes. Animals (Basel) 2024; 14:3066. [PMID: 39518789 PMCID: PMC11545463 DOI: 10.3390/ani14213066] [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: 08/12/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
The present study aimed to investigate whether supplementation of modified lysophospholipids (LPLs) in the diet of broiler breeders can benefit their offspring. A total of 264 49-week-old breeders (Ross 308) were allocated and fed based on a 2 × 2 factorial arrangement with two levels of dietary energy (normal energy = 2800 kcal/kg and low energy = 2760 kcal/kg) and two LPL levels (0 and 0.5 g/kg) for periods of 8 and 12 weeks. The offspring were assessed for growth performance, serum parameters, hepatic antioxidative capability, and expression of genes involved in liver β-oxidation at 7 days old. The LPL inclusion improved (p < 0.01) average body weight (ABW), average daily gain (ADG), and feed conversion ratio (FCR). The offspring of 61-week-old breeders fed with LPL exhibited reduced serum triglyceride levels (p < 0.01) but an increase in hepatic glutathione peroxidase (p < 0.05). The LPL increased (p < 0.001) the mRNA expression of the PGC-1α gene in the liver. Supplementing LPL in low-energy diets resulted in higher FABP1 gene expression (p < 0.05) in the intestine. In conclusion, LPL supplementation in the breeders' diet improved offspring performance by enhancing fatty acid absorption, hepatic indices, and the expression of genes involved in liver β-oxidation.
Collapse
Affiliation(s)
- Mohammad Sedghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Fatemeh Javanmard
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Anvar Amoozmehr
- Department of Animal and Poultry Nutrition, Faculty of Animal Science, Gorgan University of Agricultural Sciences and Natural Resources, Shahid Beheshti Ave, Gorgan 49138-15739, Iran;
| | - Saeid Zamany
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Ishmael Mohammadi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran; (F.J.); (S.Z.); (I.M.)
| | - Woo Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; (W.K.); (V.S.R.C.)
| | | |
Collapse
|
3
|
Guan S, Xu Z, Yang T, Zhang Y, Zheng Y, Chen T, Liu H, Zhou J. Identifying potential targets for preventing cancer progression through the PLA2G1B recombinant protein using bioinformatics and machine learning methods. Int J Biol Macromol 2024; 276:133918. [PMID: 39019365 DOI: 10.1016/j.ijbiomac.2024.133918] [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: 05/28/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/19/2024]
Abstract
Lung cancer is the deadliest and most aggressive malignancy in the world. Preventing cancer is crucial. Therefore, the new molecular targets have laid the foundation for molecular diagnosis and targeted therapy of lung cancer. PLA2G1B plays a key role in lipid metabolism and inflammation. PLA2G1B has selective substrate specificity. In this paper, the recombinant protein molecular structure of PLA2G1B was studied and novel therapeutic interventions were designed to disrupt PLA2G1B activity and impede tumor growth by targeting specific regions or residues in its structure. Construct protein-protein interaction networks and core genes using R's "STRING" program. LASSO, SVM-RFE and RF algorithms identified important genes associated with lung cancer. 282 deg were identified. Enrichment analysis showed that these genes were mainly related to adhesion and neuroactive ligand-receptor interaction pathways. PLA2G1B was subsequently identified as developing a preventative feature. GSEA showed that PLA2G1B is closely related to α-linolenic acid metabolism. Through the analysis of LASSO, SVM-RFE and RF algorithms, we found that PLA2G1B gene may be a preventive gene for lung cancer.
Collapse
Affiliation(s)
- Shuhong Guan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | | | | | | | | | | | - Huimin Liu
- Nanjing Medical University, Nanjing 211166, China
| | - Jun Zhou
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou 213000, China.
| |
Collapse
|
4
|
Haller AM, Wolfkiel PR, Jaeschke A, Hui DY. Inactivation of Group 1B Phospholipase A 2 Enhances Disease Recovery and Reduces Experimental Colitis in Mice. Int J Mol Sci 2023; 24:16155. [PMID: 38003345 PMCID: PMC10671771 DOI: 10.3390/ijms242216155] [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: 10/20/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Phospholipase A2 (PLA2) enzymes influence inflammatory bowel disease in both positive and negative manners depending on the type of PLA2 that is expressed. This study explored the influence of the abundantly expressed Group 1B PLA2 (PLA2G1B) on ulcerative colitis. Wild-type C57BL/6J mice and Pla2g1b-/- mice were treated with dextran sulfate sodium (DSS) for 5 days to induce epithelial injury, followed by another 5 days without DSS for recovery. The Pla2g1b-/- mice displayed significantly less body weight loss, colitis pathology, and disease activity indexes compared to the wild-type mice. The differences in colitis were not due to differences in the colonic lysophospholipid levels, but higher numbers of stem and progenitor cells were found in the intestines of Pla2g1b-/- mice compared to the wild-type mice. The DSS-treated Pla2g1b-/- mice also showed higher expressions of genes that are responsible for epithelial repair and lower expressions of proinflammatory cytokine genes in the colon, as well as reduced inflammatory cytokine levels in the plasma. In vitro experiments revealed the PLA2G1B stimulation of inflammatory cytokine expression by myeloid cells. PLA2G1B inactivation protects against DSS-induced colitis in mice by increasing the intestinal stem cell reservoir for epithelial repair and reducing myeloid cell inflammation in the diseased colon. Thus, PLA2G1B may be a target for colitis management.
Collapse
Affiliation(s)
- April M. Haller
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (A.M.H.); (A.J.)
| | - Patrick R. Wolfkiel
- Molecular Genetics, Biochemistry and Microbiology Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Anja Jaeschke
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (A.M.H.); (A.J.)
| | - David Y. Hui
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (A.M.H.); (A.J.)
| |
Collapse
|
5
|
Murakami M, Sato H, Taketomi Y. Modulation of immunity by the secreted phospholipase A 2 family. Immunol Rev 2023; 317:42-70. [PMID: 37035998 DOI: 10.1111/imr.13205] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Among the phospholipase A2 (PLA2 ) superfamily, which typically catalyzes the sn-2 hydrolysis of phospholipids to yield fatty acids and lysophospholipids, the secreted PLA2 (sPLA2 ) family contains 11 isoforms in mammals. Individual sPLA2 s have unique enzymatic specificity toward fatty acids and polar heads of phospholipid substrates and display distinct tissue/cellular distributions, suggesting their distinct physiological functions. Recent studies using knockout and/or transgenic mice for a full set of sPLA2 s have revealed their roles in modulation of immunity and related disorders. Application of mass spectrometric lipidomics to these mice has enabled to identify target substrates and products of individual sPLA2 s in given tissue microenvironments. sPLA2 s hydrolyze not only phospholipids in the plasma membrane of activated, damaged or dying mammalian cells, but also extracellular phospholipids such as those in extracellular vesicles, microbe membranes, lipoproteins, surfactants, and dietary phospholipids, thereby exacerbating or ameliorating various diseases. The actions of sPLA2 s are dependent on, or independent of, the generation of fatty acid- or lysophospholipid-derived lipid mediators according to the pathophysiological contexts. In this review, we make an overview of our current understanding of the roles of individual sPLA2 s in various immune responses and associated diseases.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Hiroyasu Sato
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Khan SR, Rost H, Cox B, Razani B, Alexeeff S, Wheeler MB, Gunderson EP. Heterogeneity in Early Postpartum Metabolic Profiles Among Women with GDM Who Progressed to Type 2 Diabetes During 10-Year Follow-Up: The SWIFT Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.13.23291346. [PMID: 37398098 PMCID: PMC10312884 DOI: 10.1101/2023.06.13.23291346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
GDM is a strong risk factor for progression to T2D after pregnancy. Although both GDM and T2D exhibit heterogeneity, the link between the distinct heterogeneity of GDM and incident T2D has not been established. Herein, we evaluate early postpartum profiles of women with recent GDM who later developed incident T2D using a soft clustering method, followed by the integration of both clinical phenotypic variables and metabolomics to characterize these heterogeneous clusters/groups clinically and their molecular mechanisms. We identified three clusters based on two indices of glucose homeostasis at 6-9 weeks postpartum - HOMA-IR and HOMA-B among women who developed incident T2D during the 12-year follow-up. The clusters were classified as follows: pancreatic beta-cell dysfunction group (cluster-1), insulin resistant group (cluster-3), and a combination of both phenomena (cluster-2) comprising the majority of T2D. We also identified postnatal blood test parameters to distinguish the three clusters for clinical testing. Moreover, we compared these three clusters in their metabolomics profiles at the early stage of the disease to identify the mechanistic insights. A significantly higher concentration of a metabolite at the early stage of a T2D cluster than other clusters indicates its essentiality for the particular disease character. As such, the early-stage characters of T2D cluster-1 pathology include a higher concentration of sphingolipids, acyl-alkyl phosphatidylcholines, lysophosphatidylcholines, and glycine, indicating their essentiality for pancreatic beta-cell function. In contrast, the early-stage characteristics of T2D cluster-3 pathology include a higher concentration of diacyl phosphatidylcholines, acyl-carnitines, isoleucine, and glutamate, indicating their essentiality for insulin actions. Notably, all these biomolecules are found in the T2D cluster-2 with mediocre concentrations, indicating a true nature of a mixed group. In conclusion, we have deconstructed incident T2D heterogeneity and identified three clusters with their clinical testing procedures and molecular mechanisms. This information will aid in adopting proper interventions using a precision medicine approach.
Collapse
Affiliation(s)
- Saifur R Khan
- Department of Cardiology, University of Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, PA, USA
- Departments of Physiology and Medicine, University of Toronto, Ontario, Canada
| | - Hannes Rost
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Brian Cox
- Department of Obstetrics and Gynaecology, University of Toronto, Ontario, Canada
| | - Babak Razani
- Department of Cardiology, University of Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, PA, USA
| | - Stacey Alexeeff
- Kaiser Permanente Northern California, Division of Research, Oakland, CA
| | - Michael B Wheeler
- Departments of Physiology and Medicine, University of Toronto, Ontario, Canada
| | - Erica P Gunderson
- Kaiser Permanente Northern California, Division of Research, Oakland, CA
- Kaiser Permanente Bernard J. Tyson School of Medicine, Department of Health Systems Science, Pasadena, CA
| |
Collapse
|
7
|
Zhong YH, Liang J, Qin Q, Wang YJ, Peng YM, Zhang T, Liu FY, Zhang XY, He JW, Zhang SW, Zhong GY, Huang HL, Zeng JX. The activities and mechanisms of intestinal microbiota metabolites of TCM herbal ingredients could be illustrated by a strategy integrating spectrum-effects, network pharmacology, metabolomics and molecular docking analysis: Platycodin D as an example. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154831. [PMID: 37094423 DOI: 10.1016/j.phymed.2023.154831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/25/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND The intestinal microbiota plays a key role in understanding the mechanism of traditional Chinese medicine (TCM), as it could transform the herbal ingredients to metabolites with higher bioavailability and activity comparing to their prototypes. Nevertheless, the study of the activity and mechanism of microbiota metabolites reported by the published literature still lacks viable ways. Hence a new strategy is proposed to solve this issue. PURPOSE A new strategy to study the activity and mechanism of intestinal microbiota metabolites of TCM herbal ingredients by integrating spectrum-effect relationship, network pharmacology, metabolomics analysis and molecular docking together was developed and proposed. METHOD Platycodin D (PD) and its microbiota metabolites with antitussive and expectorant effect were selected as an example for demonstration. First, the PD and its microbiota metabolites with important contribution to antitussive and/or expectorant effects were screened through spectrum-effect relationship analysis. Second, network pharmacology and metabolomics analysis were integrated to identify the upstream key targets of PD and its microbiota metabolites as well as the downstream endogenous metabolites. Finally, the active forms of PD were further confirmed by molecular docking. RESULTS Results showed that PD was an active ingredient with antitussive and/or expectorant effects, and the active forms of PD were its microbiota metabolites: 3-O-β-d-glucopyranosyl platycodigenin, 3-O-β-d-glucopyranosyl isoplatycodigenin, 7‑hydroxyl-3-O-β-d-glucopyranosyl platycodigenin, platycodigenin and isoplatycodigenin. In addition, those microbiota metabolites could bind the key targets of PAH, PLA2G2A, ALOX5, CYP2C9 and CYP2D6 to exert antitussive effects by regulating four metabolic pathways of phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, glycerophospholipid metabolism and linoleic acid metabolism. Similarly, they could also bind the key targets of PLA2G1B, ALOX5, CYP2C9 and CYP2D6 to exert expectorant effect by regulating two pathways of glycerophospholipid metabolism and linoleic acid metabolism. CONCLUSION The proposed strategy paves a new way for the illustration of the activities and mechanisms of TCM herbal ingredients, which is very important to reconcile the conundrums of TCM herbal ingredients with low oral bioavailability but high activity.
Collapse
Affiliation(s)
- Yuan-Han Zhong
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jian Liang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qian Qin
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yu-Jie Wang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yi-Ming Peng
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ting Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Fang-Yuan Liu
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xin-Yu Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jun-Wei He
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Shou-Wen Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Guo-Yue Zhong
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Hui-Lian Huang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jin-Xiang Zeng
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
8
|
Ewers M, Epple D, Bugert P, Rosendahl J, Witt H. Genetic analysis of pancreatic phospholipase A2 (PLA2G1B) in patients with chronic pancreatitis. Pancreatology 2022; 22:244-247. [PMID: 35031208 DOI: 10.1016/j.pan.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Genetic mutations in various pancreatic enzymes or their counteracting proteins have been linked to chronic pancreatitis. In particular, variants in the genes encoding pancreatic lipase (PNLIP) and carboxyl ester lipase (CEL) have been associated with pancreatitis. Therefore, we investigated pancreatic phospholipase A2 (PLA2G1B) as a promising candidate gene in patients with chronic pancreatitis. METHODS We analyzed all coding exons and adjacent intronic regions of PLA2G1B in 416 German patients with non-alcoholic chronic pancreatitis (NACP) and 186 control subjects by direct DNA sequencing. RESULTS We detected 2 frequent synonymous variants in exon 3: c.222T>C (p.Y74 = ) and c.294G>A (p.S98 = ). The genotype and allele frequencies of these variants were similar between patients and controls (c.222 TC: 9.6% in NACP vs. 9.7% in controls; c.222CC: 0.2% in NACP vs. 0% in controls; c.294 GA: 31.3% in NACP vs. 28.0% in controls; c.294AA: 2.4% in NACP vs. 1.1% in controls). All p-values were non-significant. In addition, we found one synonymous variant, c.138C>T (p.N46 = ) and one non-synonymous variant, c.244A>G (p.S82G), in a single case each. CONCLUSIONS Our results suggest that genetic alterations in PLA2G1B do not predispose to the development of non-alcoholic chronic pancreatitis.
Collapse
Affiliation(s)
- Maren Ewers
- Pediatric Nutritional Medicine & Else Kröner-Fresenius-Centre for Nutritional Medicine (EKFZ), Technical University Munich (TUM), Freising, Germany
| | - Denise Epple
- Pediatric Nutritional Medicine & Else Kröner-Fresenius-Centre for Nutritional Medicine (EKFZ), Technical University Munich (TUM), Freising, Germany; Department of Pediatrics, MRI, Technical University Munich (TUM), Munich, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Heidelberg University, Medical Faculty Mannheim, German Red Cross Blood Service Baden, Württemberg, Hessen, Mannheim, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin Luther University, Halle, Germany
| | - Heiko Witt
- Pediatric Nutritional Medicine & Else Kröner-Fresenius-Centre for Nutritional Medicine (EKFZ), Technical University Munich (TUM), Freising, Germany.
| |
Collapse
|
9
|
Hundal BK, Lutfi E, Sigholt T, Rosenlund G, Liland NS, Glencross B, Sissener NH. A Piece of the Puzzle-Possible Mechanisms for Why Low Dietary EPA and DHA Cause Hepatic Lipid Accumulation in Atlantic Salmon ( Salmo salar). Metabolites 2022; 12:159. [PMID: 35208233 PMCID: PMC8877222 DOI: 10.3390/metabo12020159] [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: 01/18/2022] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/23/2022] Open
Abstract
The present study aimed at elucidating the effects of graded levels of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on the hepatic metabolic health of Atlantic salmon reared in sea cages. Diets containing 10, 13, 16 and 35 g/kg EPA + DHA (designated diets 1.0, 1.3, 1.6 and 3.5, respectively) were fed in triplicate through a full production cycle from an average starting weight of 275 g to slaughter size (~5 kg). Feeding low dietary EPA + DHA altered the hepatic energy metabolism, evidenced by reductions in tricarboxylic acid cycle intermediates originating from β-oxidation, which was compensated by elevated activity in alternative energy pathways (pentose phosphate pathway, branched chain amino acid catabolism and creatine metabolism). Increases in various acylcarnitines in the liver supported this and indicates issues with lipid metabolism (mitochondrial β-oxidation). Problems using lipids for energy in the lower EPA + DHA groups line up well with observed increases in liver lipids in these fish. It also aligns with the growth data, where fish fed the highest EPA + DHA grew better than the other groups. The study showed that diets 1.0 and 1.3 were insufficient for maintaining good liver metabolic health. However, diet 3.5 was significantly better than diet 1.6, indicating that diet 1.6 might also be suboptimal.
Collapse
Affiliation(s)
- Bjørg Kristine Hundal
- Department of Feed and Nutrition, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway; (N.S.L.); (N.H.S.)
| | - Esmail Lutfi
- Department of Nutrition and Feed Technology, Norwegian Institute of Food, Fisheries and Aquaculture Research (Nofima), P.O. Box 210, 1431 Ås, Norway;
| | | | - Grethe Rosenlund
- Skretting Aquaculture Research Centre, P.O. Box 48, 4001 Stavanger, Norway;
| | - Nina Sylvia Liland
- Department of Feed and Nutrition, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway; (N.S.L.); (N.H.S.)
| | - Brett Glencross
- Institute of Aquaculture, University of Stirling, Stirling FK9 4LA, UK;
| | - Nini Hedberg Sissener
- Department of Feed and Nutrition, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway; (N.S.L.); (N.H.S.)
| |
Collapse
|
10
|
Watkins OC, Selvam P, Appukuttan Pillai R, Cracknell-Hazra VKB, Yong HEJ, Sharma N, Cazenave-Gassiot A, Bendt AK, Godfrey KM, Lewis RM, Wenk MR, Chan SY. Placental 13C-DHA metabolism and relationship with maternal BMI, glycemia and birthweight. Mol Med 2021; 27:84. [PMID: 34362294 PMCID: PMC8349043 DOI: 10.1186/s10020-021-00344-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fetal docosahexaenoic acid (DHA) supply relies on preferential transplacental transfer, which is regulated by placental DHA lipid metabolism. Maternal hyperglycemia and obesity associate with higher birthweight and fetal DHA insufficiency but the role of placental DHA metabolism is unclear. METHODS Explants from 17 term placenta were incubated with 13C-labeled DHA for 48 h, at 5 or 10 mmol/L glucose treatment, and the production of 17 individual newly synthesized 13C-DHA labeled lipids quantified by liquid chromatography mass spectrometry. RESULTS Maternal BMI positively associated with 13C-DHA-labeled diacylglycerols, triacylglycerols, lysophospholipids, phosphatidylcholine and phosphatidylethanolamine plasmalogens, while maternal fasting glycemia positively associated with five 13C-DHA triacylglycerols. In turn, 13C-DHA-labeled phospholipids and triacylglycerols positively associated with birthweight centile. In-vitro glucose treatment increased most 13C-DHA-lipids, but decreased 13C-DHA phosphatidylethanolamine plasmalogens. However, with increasing maternal BMI, the magnitude of the glucose treatment induced increase in 13C-DHA phosphatidylcholine and 13C-DHA lysophospholipids was curtailed, with further decline in 13C-DHA phosphatidylethanolamine plasmalogens. Conversely, with increasing birthweight centile glucose treatment induced increases in 13C-DHA triacylglycerols were exaggerated, while glucose treatment induced decreases in 13C-DHA phosphatidylethanolamine plasmalogens were diminished. CONCLUSIONS Maternal BMI and glycemia increased the production of different placental DHA lipids implying impact on different metabolic pathways. Glucose-induced elevation in placental DHA metabolism is moderated with higher maternal BMI. In turn, findings of associations between many DHA lipids with birthweight suggest that BMI and glycemia promote fetal growth partly through changes in placental DHA metabolism.
Collapse
Affiliation(s)
- Oliver C Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Victoria K B Cracknell-Hazra
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anne K Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Rohan M Lewis
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
11
|
Kang S, Lee AY, Park SY, Liu KH, Im DS. O-1602 Promotes Hepatic Steatosis through GPR55 and PI3 Kinase/Akt/SREBP-1c Signaling in Mice. Int J Mol Sci 2021; 22:3091. [PMID: 33803038 PMCID: PMC8003045 DOI: 10.3390/ijms22063091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 11/30/2022] Open
Abstract
Non-alcoholic fatty liver disease is recognized as the leading cause of chronic liver disease. Overnutrition and obesity are associated with hepatic steatosis. G protein-coupled receptor 55 (GPR55) has not been extensively studied in hepatic steatosis, although its endogenous ligands have been implicated in liver disease progression. Therefore, the functions of GPR55 were investigated in Hep3B human hepatoma cells and mice fed high-fat diets. O-1602, the most potent agonist of GPR55, induced lipid accumulation in hepatocytes, which was reversed by treatment with CID16020046, an antagonist of GPR55. O-1602 also induced intracellular calcium rise in Hep3B cells in a GPR55-independent manner. O-1602-induced lipid accumulation was dependent on the PI3 kinase/Akt/SREBP-1c signaling cascade. Furthermore, we found increased levels of lysophosphatidylinositol species of 16:0, 18:0, 18:1, 18:2, 20:1, and 20:2 in the livers of mice fed a high-fat diet for 4 weeks. One-week treatment with CID16020046 suppressed high-fat diet-induced lipid accumulation and O-1602-induced increase of serum triglyceride levels in vivo. Therefore, the present data suggest the pro-steatotic function of GPR55 signaling in hepatocytes and provide a potential therapeutic target for non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Saeromi Kang
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (S.K.); (A.-Y.L.)
| | - Ae-Yeon Lee
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (S.K.); (A.-Y.L.)
| | - So-Young Park
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (K.-H.L.)
| | - Kwang-Hyeon Liu
- BK21 FOUR KNU Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (S.-Y.P.); (K.-H.L.)
| | - Dong-Soon Im
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (S.K.); (A.-Y.L.)
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
12
|
Braga AA, Bortolin RH, Graciano-Saldarriaga ME, Hirata TD, Cerda A, de Freitas RC, Lin-Wang HT, Borges JB, França JI, Masi LN, Curi R, Pithon-Curi TC, Sampaio MF, Castro LR, Bastos GM, Hirata RD, Hirata MH. High serum miR-421 is associated with metabolic dysregulation and inflammation in patients with metabolic syndrome. Epigenomics 2021; 13:423-436. [PMID: 33678000 DOI: 10.2217/epi-2020-0247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: To explore the association of circulating miRNAs with adiposity, metabolic status and inflammatory biomarkers in patients with metabolic syndrome (MetS). Methods: Serum levels of 372 miRNAs were measured in patients with (n = 6) and without MetS (n = 6) by quantitative PCR array, and dysregulated miRNAs were validated in a larger cohort (MetS, n = 89; non-MetS, n = 144). Results: In the screening study, seven miRNAs were dysregulated in patients with MetS, and miR-421 remained increased in the validation study. miR-421 was associated with a high risk of MetS and insulin resistance and hypertension and correlated with glycated hemoglobin, triacylglycerols, high-sensitivity CRP, IL-6, resistin and adiponectin (p < 0.05). Conclusion: Circulating miR-421 is a potential biomarker for insulin resistance, metabolic dysregulation and inflammatory status in patients with MetS.
Collapse
Affiliation(s)
- Aécio A Braga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Raul H Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Magda E Graciano-Saldarriaga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Thiago Dc Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, CEMT-BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Renata Cc de Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Hui T Lin-Wang
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - Jessica B Borges
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - João Id França
- Laboratory of Epidemiology and Statistics, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - Laureane N Masi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Tania C Pithon-Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Marcelo F Sampaio
- Medical Clinic Division, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Cardiology, Real e Benemerita Associação Portuguesa de Beneficiência, Sao Paulo 01323-001, Brazil
| | - Lara R Castro
- Medical Clinic Division, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Cardiology, Real e Benemerita Associação Portuguesa de Beneficiência, Sao Paulo 01323-001, Brazil
| | - Gisele M Bastos
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Teaching and Research, Real e Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | - Rosario Dc Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Mario H Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
13
|
Fondevila MF, Fernandez U, Gonzalez-Rellan MJ, Da Silva Lima N, Buque X, Gonzalez-Rodriguez A, Alonso C, Iruarrizaga-Lejarreta M, Delgado TC, Varela-Rey M, Senra A, Garcia-Outeiral V, Novoa E, Iglesias C, Porteiro B, Beiroa D, Folgueira C, Tojo M, Torres JL, Hernández-Cosido L, Blanco Ó, Arab JP, Barrera F, Guallar D, Fidalgo M, López M, Dieguez C, Marcos M, Martinez-Chantar ML, Arrese M, Garcia-Monzon C, Mato JM, Aspichueta P, Nogueiras R. The L-α-Lysophosphatidylinositol/G Protein-Coupled Receptor 55 System Induces the Development of Nonalcoholic Steatosis and Steatohepatitis. Hepatology 2021; 73:606-624. [PMID: 32329085 PMCID: PMC7894478 DOI: 10.1002/hep.31290] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/24/2020] [Accepted: 04/05/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS G protein-coupled receptor (GPR) 55 is a putative cannabinoid receptor, and l-α-lysophosphatidylinositol (LPI) is its only known endogenous ligand. Although GPR55 has been linked to energy homeostasis in different organs, its specific role in lipid metabolism in the liver and its contribution to the pathophysiology of nonalcoholic fatty liver disease (NAFLD) remains unknown. APPROACH AND RESULTS We measured (1) GPR55 expression in the liver of patients with NAFLD compared with individuals without obesity and without liver disease, as well as animal models with steatosis and nonalcoholic steatohepatitis (NASH), and (2) the effects of LPI and genetic disruption of GPR55 in mice, human hepatocytes, and human hepatic stellate cells. Notably, we found that circulating LPI and liver expression of GPR55 were up-regulated in patients with NASH. LPI induced adenosine monophosphate-activated protein kinase activation of acetyl-coenzyme A carboxylase (ACC) and increased lipid content in human hepatocytes and in the liver of treated mice by inducing de novo lipogenesis and decreasing β-oxidation. The inhibition of GPR55 and ACCα blocked the effects of LPI, and the in vivo knockdown of GPR55 was sufficient to improve liver damage in mice fed a high-fat diet and in mice fed a methionine-choline-deficient diet. Finally, LPI promoted the initiation of hepatic stellate cell activation by stimulating GPR55 and activation of ACC. CONCLUSIONS The LPI/GPR55 system plays a role in the development of NAFLD and NASH by activating ACC.
Collapse
Affiliation(s)
- Marcos F Fondevila
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain
| | - Uxia Fernandez
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Maria J Gonzalez-Rellan
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Natalia Da Silva Lima
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Xabier Buque
- Department of PhysiologyUniversity of the Basque Country UPV/EHULeioaSpain.,Biocruces Bizkaia Health Research InstituteBarakaldoSpain
| | - Agueda Gonzalez-Rodriguez
- Liver Research UnitSanta Cristina University HospitalInstituto de Investigación Sanitaria PrincesaMadridSpain
| | | | | | - Teresa C Delgado
- Liver Disease LaboratoryCenter for Cooperative Research in BiosciencesBasque Research and Technology Alliance-Centro de Enfermedades Hepáticas y DigestivasCentro de Investigación Biomédica en RedDerioSpain
| | - Marta Varela-Rey
- Liver Disease LaboratoryCenter for Cooperative Research in BiosciencesBasque Research and Technology Alliance-Centro de Enfermedades Hepáticas y DigestivasCentro de Investigación Biomédica en RedDerioSpain
| | - Ana Senra
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Vera Garcia-Outeiral
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Eva Novoa
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Cristina Iglesias
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Begoña Porteiro
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain
| | - Daniel Beiroa
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain
| | - Cintia Folgueira
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Marta Tojo
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Jorge L Torres
- Department of Internal MedicineUniversity Hospital of Salamanca-Institute of Biomedical Research of SalamancaUniversity of SalamancaSalamancaSpain
| | - Lourdes Hernández-Cosido
- Department of General and Gastrointestinal SurgeryUniversity Hospital of Salamanca-Institute of Biomedical Research of SalamancaUniversity of SalamancaSalamancaSpain
| | - Óscar Blanco
- Department of PathologyUniversity Hospital of Salamanca-Institute of Biomedical Research of SalamancaUniversity of SalamancaSalamancaSpain
| | - Juan Pablo Arab
- Departament of GastroenterologyEscuela de MedicinaPontificia Universidad Católica de Chile, Santiago, ChileChile and Centro de Envejecimiento y Regeneración (CARE) Facultad de Ciencias Biológicaspontificia Universidad Católica de ChileSantiagoChile
| | - Francisco Barrera
- Departament of GastroenterologyEscuela de MedicinaPontificia Universidad Católica de Chile, Santiago, ChileChile and Centro de Envejecimiento y Regeneración (CARE) Facultad de Ciencias Biológicaspontificia Universidad Católica de ChileSantiagoChile
| | - Diana Guallar
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Miguel Fidalgo
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain
| | - Miguel López
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain
| | - Carlos Dieguez
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain
| | - Miguel Marcos
- Department of Internal MedicineUniversity Hospital of Salamanca-Institute of Biomedical Research of SalamancaUniversity of SalamancaSalamancaSpain
| | - Maria L Martinez-Chantar
- Liver Disease LaboratoryCenter for Cooperative Research in BiosciencesBasque Research and Technology Alliance-Centro de Enfermedades Hepáticas y DigestivasCentro de Investigación Biomédica en RedDerioSpain
| | - Marco Arrese
- Departament of GastroenterologyEscuela de MedicinaPontificia Universidad Católica de Chile, Santiago, ChileChile and Centro de Envejecimiento y Regeneración (CARE) Facultad de Ciencias Biológicaspontificia Universidad Católica de ChileSantiagoChile
| | - Carmelo Garcia-Monzon
- Liver Research UnitSanta Cristina University HospitalInstituto de Investigación Sanitaria PrincesaMadridSpain
| | - Jose M Mato
- Liver Disease LaboratoryCenter for Cooperative Research in BiosciencesBasque Research and Technology Alliance-Centro de Enfermedades Hepáticas y DigestivasCentro de Investigación Biomédica en RedDerioSpain.,Liver Metabolism LaboratoryCenter for Cooperative Research in Biosciences, Basque Research and Technology Alliance-Centro de Enfermedades Hepáticas y DigestivasCentro de Investigación Biomédica en RedDerioSpain
| | - Patricia Aspichueta
- Department of PhysiologyUniversity of the Basque Country UPV/EHULeioaSpain.,Biocruces Bizkaia Health Research InstituteBarakaldoSpain
| | - Ruben Nogueiras
- Department of PhysiologyResearch Centre of Molecular Medicine and Chronic DiseasesUniversity of Santiago de Compostela-Instituto de Investigación SanitariaSantiago de CompostelaSpain.,Centro de Fisiopatología de la Obesidad y NutriciónCentro de Investigación Biomédica en RedSantiago de CompostelaSpain.,Galician Agency of Innovation (GAIN)Xunta de GaliciaSantiago de CompostelaSpain
| |
Collapse
|
14
|
Kuefner MS. Secretory Phospholipase A2s in Insulin Resistance and Metabolism. Front Endocrinol (Lausanne) 2021; 12:732726. [PMID: 34512555 PMCID: PMC8429832 DOI: 10.3389/fendo.2021.732726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
The phospholipases A2 (PLA2) superfamily encompasses enzymes commonly found in mammalian tissues and snake venom. Many of these enzymes have unique tissue distribution, function, and substrate specificity suggesting distinct biological roles. In the past, much of the research on secretory PLA2s has analyzed their roles in inflammation, anti-bacterial actions, and atherosclerosis. In recent studies utilizing a variety of mouse models, pancreatic islets, and clinical trials, a role for many of these enzymes in the control of metabolism and insulin action has been revealed. In this review, this research, and the unique contributions of the PLA2 enzymes in insulin resistance and metabolism.
Collapse
|
15
|
Fu L, Jiang Y, Wang C, Mei M, Zhou Z, Jiang Y, Song H, Ding X. A Genome-Wide Association Study on Feed Efficiency Related Traits in Landrace Pigs. Front Genet 2020; 11:692. [PMID: 32719719 PMCID: PMC7350416 DOI: 10.3389/fgene.2020.00692] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Feed efficiency (FE) traits in pigs are of utmost economic importance. Genetic improvement of FE related traits in pigs might significantly reduce production cost and energy consumption. Hence, our study aimed at identifying SNPs and candidate genes associated with FE related traits, including feed conversion ratio (FCR), average daily gain (ADG), average daily feed intake (ADFI), and residual feed intake (RFI). A genome-wide association study (GWAS) was performed for the four FE related traits in 296 Landrace pigs genotyped with PorcineSNP50 BeadChip. Two different single-trait methods, single SNP linear model GWAS (LM-GWAS) and single-step GWAS (ssGWAS), were implemented. Our results showed that the two methods showed high consistency with respect to SNP identification. A total of 32 common significant SNPs associated with the four FE related traits were identified. Bioinformatics analysis revealed eight common QTL regions, of which three QTL regions related to ADFI and RFI traits were overlapped. Gene ontology analysis revealed six common candidate genes (PRELID2, GPER1, PDX1, TEX2, PLCL2, ICAM2) relevant for the four FE related traits. These genes are involved in the processes of fat synthesis and decomposition, lipid transport process, insulin metabolism, among others. Our results provide, new insights into the genetic mechanisms and candidate function genes of FE related traits in pigs. However, further investigations to validate these results are warranted.
Collapse
Affiliation(s)
- Lu Fu
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yao Jiang
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chonglong Wang
- Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Mengran Mei
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ziwen Zhou
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifan Jiang
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hailiang Song
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiangdong Ding
- National Engineering Laboratory for Animal Breeding, Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
16
|
Sato H, Taketomi Y, Miki Y, Murase R, Yamamoto K, Murakami M. Secreted Phospholipase PLA2G2D Contributes to Metabolic Health by Mobilizing ω3 Polyunsaturated Fatty Acids in WAT. Cell Rep 2020; 31:107579. [DOI: 10.1016/j.celrep.2020.107579] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 11/18/2019] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
|
17
|
Nishiumi S, Izumi Y, Kobayashi T, Yoshida M. Possible Involvement of Lipids in the Effectiveness of Kombu in Individuals with Abnormally High Serum Triglyceride Levels. J Nutr Sci Vitaminol (Tokyo) 2020; 66:185-190. [PMID: 32350180 DOI: 10.3177/jnsv.66.185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In Japan, Kombu (Laminaria japonica), which is a type of seaweed, is considered to be a foodstuff with health-promoting benefits, and Japanese people actively incorporate Kombu into their diets. Previously, we reported that the frequent intake of Kombu reduced the serum triglyceride levels of subjects with abnormally high serum triglyceride levels. In the current human study, we performed metabolomic analysis of serum lipids, and then the molecular species profiles of phosphatidylcholines (PC), phosphatidylethanolamines (PE), lysophosphatidylcholines (LPC), lysophosphatidylethanolamines (LPE), and free fatty acids (FFA) were evaluated. As a result, it was found that there were no marked differences between the lipid profiles obtained before and after the intake of Kombu for 4 wk in all subjects. In the subjects with abnormal serum triglyceride levels, the intake of Kombu improved the subjects' molecular species profiles in terms of their serum levels of the diacyl and acyl forms of PC, PE, LPC, and LPE, and FFA. Furthermore, the intake of Kombu also tended to increase the serum levels of both the plasmanyl and plasmenyl forms of PC and PE in these subjects. The lipid alterations observed in our study might be related to the functionality of Kombu. Furthermore, it is important to evaluate the quality of lipids as well as the quantity of lipids in various types of research, including food functionality studies.
Collapse
Affiliation(s)
- Shin Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
- Department of Omics Medicine, Hyogo College of Medicine
| | - Yoshihiro Izumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Masaru Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
- Division of Metabolomics Research, Department of Internal Medicine Related, Kobe University Graduate School of Medicine
- AMED-CREST, AMED
| |
Collapse
|
18
|
Adamska-Patruno E, Samczuk P, Ciborowski M, Godzien J, Pietrowska K, Bauer W, Gorska M, Barbas C, Kretowski A. Metabolomics Reveal Altered Postprandial Lipid Metabolism After a High-Carbohydrate Meal in Men at High Genetic Risk of Diabetes. J Nutr 2019; 149:915-922. [PMID: 31049566 DOI: 10.1093/jn/nxz024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/17/2018] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The transcription factor 7-like 2 (TCF7L2) gene confers one of the strongest genetic predispositions to type 2 diabetes, but diabetes development can be modified by diet. OBJECTIVE The aim of our study was to evaluate postprandial metabolic alterations in healthy men with a high genetic risk of diabetes, after two meals with varying macronutrient content. METHODS The study was conducted in 21 homozygous nondiabetic men carrying the high-risk (HR, n = 8, age: 31.2 ± 6.3 y, body mass index (BMI, kg/m2) 28.5 ± 8.1) or low-risk (LR, n = 13, age: 35.2 ± 10.3 y, BMI: 28.1 ± 6.4) genotypes at the rs7901695 locus. During two meal challenge test visits subjects received standardized isocaloric (450 kcal) liquid meals: high-carbohydrate (HC, carbohydrates: 89% of energy) and normo-carbohydrate (NC, carbohydrates: 45% of energy). Fasting (0 min) and postprandial (30, 60, 120, 180 min) plasma samples were analyzed for metabolite profiles through untargeted metabolomics. Metabolic fingerprinting was performed on an ultra-high-performance liquid chromatography (UHPLC) system connected to an iFunnel quadrupole-time-of-flight (Q-TOF) mass spectrometer. RESULTS In HR-genotype men, after the intake of an HC-meal, we noted a significantly lower area under the curves (AUCs) of postprandial plasma concentrations of most of the phospholipids (-37% to -53%, variable importance in the projection (VIP) = 1.2-1.5), lysophospholipids (-29% to -86%, VIP = 1.1-2.6), sphingolipids (-32% to -47%, VIP = 1.1-1.3), as well as arachidonic (-36%, VIP = 1.4) and oleic (-63%, VIP = 1.3) acids, their metabolites: keto- and hydoxy-fatty acids (-38% to -78%, VIP = 1.3-2.5), leukotrienes (-65% to -83%, VIP = 1.4-2.2), uric acid (-59%, VIP = 1.5), and pyroglutamic acid (-65%, VIP = 1.8). The AUCs of postprandial sphingosine concentrations were higher (125-832%, VIP = 1.9-3.2) after the NC-meal, AUCs of acylcarnitines were lower (-21% to -61%, VIP = 1.1-2.4), and AUCs of fatty acid amides were higher (51-508%, VIP = 1.7-3.1) after the intake of both meals. CONCLUSIONS In nondiabetic men carrying the TCF7L2 HR genotype, subtle but detectable modifications in intermediate lipid metabolism are induced by an HC-meal. This trial was registered at www.clinicaltrials.gov as NCT03792685.
Collapse
Affiliation(s)
| | - Paulina Samczuk
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Godzien
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.,Center for Metabolomics and Bioanalysis (CEMBIO), Universidad CEU San Pablo, Madrid, Spain
| | - Karolina Pietrowska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Coral Barbas
- Center for Metabolomics and Bioanalysis (CEMBIO), Universidad CEU San Pablo, Madrid, Spain
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
19
|
Kim DH, Han SM, Choi YS, Kang HK, Lee HG, Lee KW. Effects of Dietary Bee Venom on Serum Characteristic, Antioxidant
Activity and Liver Fatty Acid Composition in Broiler Chickens. ACTA ACUST UNITED AC 2019. [DOI: 10.5536/kjps.2019.46.1.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
20
|
Li N, Zhao X, You S. Identification of key regulators of pancreatic ductal adenocarcinoma using bioinformatics analysis of microarray data. Medicine (Baltimore) 2019; 98:e14074. [PMID: 30633213 PMCID: PMC6336631 DOI: 10.1097/md.0000000000014074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer, and its etiology remains largely unknown. This study aimed to screen a panel of key genes and to identify their potential impact on the molecular pathways associated with the development of PDAC. Four gene expression profiles, GSE28735, GSE15471, GSE102238, and GSE43795, were downloaded from the Gene Expression Omnibus (GEO) database. The intersection of the differentially expressed genes (DEGs) in each dataset was obtained using Venn analysis. Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis were subsequently carried out. To screen for hub genes, a protein-protein interaction (PPI) network was constructed.The intersection of the DEGs revealed 7 upregulated and 9 downregulated genes. Upon relaxation of the selection criteria, 58 upregulated and 32 downregulated DEGs were identified. The top 5 biological processes identified by GO analysis involved peptide cross-linking, extracellular matrix (ECM) disassembly, regulation of the fibroblast growth factor receptor signaling pathway, mesoderm morphogenesis, and lipid digestion. The results of KEGG analysis revealed that the DEGs were significantly enriched in pathways involved in protein digestion and absorption, ECM-receptor interaction, pancreatic secretion, and fat digestion and absorption. The top ten hub genes were identified based on the PPI network.In conclusion, the identified hub genes may contribute to the elucidation of the underlying molecular mechanisms of PDAC and serve as promising candidates that can be utilized for the early diagnosis and prognostic prediction of PDAC. However, further experimental validation is required to confirm these results.
Collapse
Affiliation(s)
- Nan Li
- Department of General Surgery, Tianjin Medical University General Hospital
| | - Xin Zhao
- Tianjin Medical University, Tianjin, China
| | - Shengyi You
- Department of General Surgery, Tianjin Medical University General Hospital
| |
Collapse
|
21
|
Entwistle LJ, Pelly VS, Coomes SM, Kannan Y, Perez-Lloret J, Czieso S, Silva Dos Santos M, MacRae JI, Collinson L, Sesay A, Nikolov N, Metidji A, Helmby H, Hui DY, Wilson MS. Epithelial-Cell-Derived Phospholipase A 2 Group 1B Is an Endogenous Anthelmintic. Cell Host Microbe 2018; 22:484-493.e5. [PMID: 29024642 PMCID: PMC5644720 DOI: 10.1016/j.chom.2017.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 07/05/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023]
Abstract
Immunity to intestinal helminth infections has been well studied, but the mechanism of helminth killing prior to expulsion remains unclear. Here we identify epithelial-cell-derived phospholipase A2 group 1B (PLA2g1B) as a host-derived endogenous anthelmintic. PLA2g1B is elevated in resistant mice and is responsible for killing tissue-embedded larvae. Despite comparable activities of other essential type-2-dependent immune mechanisms, Pla2g1b−/− mice failed to expel the intestinal helminths Heligmosomoides polygyrus or Nippostrongylus brasiliensis. Expression of Pla2g1b by epithelial cells was dependent upon intestinal microbiota, adaptive immunity, and common-gamma chain-dependent signaling. Notably, Pla2g1b was downregulated in susceptible mice and inhibited by IL-4R-signaling in vitro, uncoupling parasite killing from expulsion mechanisms. Resistance was restored in Pla2g1b−/− mice by treating infective H. polygyrus L3 larvae with PLA2g1B, which reduced larval phospholipid abundance. These findings uncover epithelial-cell-derived Pla2g1b as an essential mediator of helminth killing, highlighting a previously overlooked mechanism of anti-helminth immunity. Pla2g1b expression correlated with resistance to intestinal helminth infection PLA2g1B is essential for resistance to intestinal helminth infection in mice PLA2g1B directly reduces phospholipid abundance in infective larvae Pla2g1b is expressed by epithelial cells and is negatively regulated by IL-4Rα
Collapse
Affiliation(s)
- Lewis J Entwistle
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Victoria S Pelly
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Stephanie M Coomes
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yashaswini Kannan
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jimena Perez-Lloret
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Stephanie Czieso
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - James I MacRae
- Metabolomics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Lucy Collinson
- Electron Microscopy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Abdul Sesay
- Advanced Sequencing Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nikolay Nikolov
- Advanced Sequencing Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Amina Metidji
- AhR Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Helena Helmby
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - David Y Hui
- Department of Pathology, Metabolic Disease Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Mark S Wilson
- Allergy and Anti-helminth Immunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Immunology Discovery, Genentech Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
22
|
Cash JG, Konaniah E, Hegde N, Kuhel DG, Watanabe M, Romick-Rosendale L, Hui DY. Therapeutic reduction of lysophospholipids in the digestive tract recapitulates the metabolic benefits of bariatric surgery and promotes diabetes remission. Mol Metab 2018; 16:55-64. [PMID: 30087032 PMCID: PMC6158127 DOI: 10.1016/j.molmet.2018.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/10/2018] [Accepted: 07/23/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Obesity and obesity-related metabolic disorders are major health problems worldwide. The most effective obesity intervention is bariatric surgery. This study tested the hypothesis that bariatric surgery alters phospholipid metabolism in the gastrointestinal tract to favor a metabolically healthy gut microbiota profile and therapeutic intervention of phospholipid metabolism in the gastrointestinal may have similar metabolic benefits. METHODS The first study compared plasma levels of the bioactive lipid metabolites lysophospholipid and trimethylamine N-oxide (TMAO) as well as gut microbiota profile in high fat/carbohydrate (HFHC) diet-fed C57BL/6 mice with or without vertical sleeve gastrectomy (VSG) and in Pla2g1b-/- mice with group 1B phospholipase A2 gene inactivation. The second study examined the effectiveness of the non-absorbable secretory phospholipase A2 inhibitor methyl indoxam to reverse hyperglycemia and hyperlipidemia in HFHC diet-fed C57BL/6 mice after diabetes onset. RESULTS Both bariatric surgery and PLA2G1B inactivation were shown to reduce lysophospholipid content in the gastrointestinal tract, resulting in resistance to HFHC diet-induced alterations of the gut microbiota, reduction of the cardiovascular risk factors hyperlipidemia and TMAO, decreased adiposity, and prevention of HFHC diet-induced diabetes. Importantly, treatment of wild type mice with methyl indoxam after HFHC diet-induced onset of hyperlipidemia and hyperglycemia effectively restored normal plasma lipid and glucose levels and replicated the metabolic benefits of VSG surgery with diabetes remission and TMAO reduction. CONCLUSION These results provided pre-clinical evidence that PLA2G1B inhibition in the digestive tract may be a viable alternative option to bariatric surgery for obesity and obesity-related cardiometabolic disorder intervention.
Collapse
Affiliation(s)
- James G Cash
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA
| | - Eddy Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA
| | - Narasimha Hegde
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA
| | - David G Kuhel
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA
| | - Miki Watanabe
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Lindsey Romick-Rosendale
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45237, USA.
| |
Collapse
|
23
|
Hui DY. Group 1B phospholipase A 2 in metabolic and inflammatory disease modulation. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:784-788. [PMID: 30003964 DOI: 10.1016/j.bbalip.2018.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/02/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022]
Abstract
The group 1B phospholipase A2 (PLA2G1B) is a secreted phospholipase that catalyzes the hydrolytic removal of the sn-2 fatty acyl moiety from phospholipids. This enzyme is synthesized most abundantly in the pancreas and is also expressed in the lung. The first part of this review article focuses on the role of pancreatic-derived PLA2G1B in mediating lipid absorption and discusses how the PLA2G1B-derived metabolic product contributes to cardiometabolic diseases, including obesity, hyperinsulinemia, hyperlipidemia, and atherosclerosis. The anti-helminth properties of PLA2G1B will also be discussed. The second part of this review will focus on PLA2G1B expressed in the lung, and in vitro data suggest that how this enzyme may modulate lung inflammation via both hydrolytic activity-dependent and -dependent mechanisms. Finally, recent studies revealing a relationship between PLA2G1B and cancer will also be discussed. This article is part of a Special Issue entitled Novel functions of phospholipase A2 Guest Editors: Makoto Murakami and Gerard Lambeau.
Collapse
Affiliation(s)
- David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; Department of Pathology, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, 2120 E. Galbraith Road, Cincinnati, OH 45237, United States.
| |
Collapse
|
24
|
Hellmuth C, Uhl O, Demmelmair H, Grunewald M, Auricchio R, Castillejo G, Korponay-Szabo IR, Polanco I, Roca M, Vriezinga SL, Werkstetter KJ, Koletzko B, Mearin ML, Kirchberg FF. The impact of human breast milk components on the infant metabolism. PLoS One 2018; 13:e0197713. [PMID: 29856767 PMCID: PMC5983411 DOI: 10.1371/journal.pone.0197713] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/27/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND & AIMS Breastfeeding is beneficial for mothers and infants. Underlying mechanisms and biochemical mediators thus need to be investigated to develop and support improved infant nutrition practices promoting the child health. We analysed the relation between maternal breast milk composition and infant metabolism. METHODS 196 pairs of mothers and infants from a European research project (PreventCD) were studied. Maternal milk samples collected at month 1 and month 4 after birth were analysed for macronutrient classes, hormone, and fatty acid (FA) content. Phospholipids, acylcarnitines, and amino acids were measured in serum samples of 4-month old infants. Associations between milk components and infant metabolites were analysed with spearman correlation and linear mixed effect models (LME). P-values were corrected for multiple testing (PLME). RESULTS Month 1 milk protein content was strongly associated with infant serum lyso-phosphatidylcholine (LPC) 14:0 (PLME = 0.009). Month 1 milk insulin was associated to infant acetylcarnitine (PLME = 0.01). There were no associations between milk protein content and serum amino acids and milk total fat content and serum polar lipids. Middle- and odd-chain FA% in breast milk at both ages were significantly related to serum LPC and sphingomyelins (SM) species in infant serum (all PLME<0.05), while FA% 20:5n-3 and 22:6n-3 percentages were significantly associated to serum LPC 22:6 (PLME = 1.91×10-4/7.93×10-5) in milk only at month 4. Other polyunsaturated fatty acids and hormones in milk showed only weak associations with infant serum metabolites. CONCLUSIONS Infant serum LPC are influenced by breast milk FA composition and, intriguingly, milk protein content in early but not late lactation. LPC 14:0, previously found positively associated with obesity risk, was the serum metabolite which was the most strongly associated to milk protein content. Thus, LPC 14:0 might be a key metabolite not only reflecting milk protein intake in infants, but also relating high protein content in milk or infant formula to childhood obesity risk.
Collapse
Affiliation(s)
- Christian Hellmuth
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| | - Olaf Uhl
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| | - Hans Demmelmair
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| | - Maria Grunewald
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| | - Renata Auricchio
- Department of Medical Translational Sciences and European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Gemma Castillejo
- Department of Pediatric Gastroenterology Unit, Hospital Universitari Sant Joan de Reus, URV, IIPV, Reus, Spain
| | - Ilma R. Korponay-Szabo
- Celiac Disease Center, Heim Pál National Pediatric Institute, Budapest, Hungary and Department of Pediatrics, University of Debrecen, Debrecen, Hungary
| | - Isabel Polanco
- Department of Pediatric Gastroenterology and Nutrition, La Paz University Hospital, Madrid, Spain
| | - María Roca
- U. Enfermedad Celiaca e Inmunopatología Digestiva, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Sabine L. Vriezinga
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Katharina J. Werkstetter
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| | - Berthold Koletzko
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
- * E-mail:
| | - M. Luisa Mearin
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Franca F. Kirchberg
- Ludwig-Maximilians-Universität München, Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children’s Hospital, University of Munich Medical Center, Munich, Germany
| |
Collapse
|
25
|
Hou S, Xu T, Xu J, Qu L, Xu Y, Chen L, Liu J. Structural basis for functional selectivity and ligand recognition revealed by crystal structures of human secreted phospholipase A 2 group IIE. Sci Rep 2017; 7:10815. [PMID: 28883454 PMCID: PMC5589937 DOI: 10.1038/s41598-017-11219-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023] Open
Abstract
Secreted phospholipases A2s (sPLA2s) are involved in various pathological conditions such as rheumatoid arthritis and cardiovascular disease. Many inhibitors were developed and studied in clinical trials, but none have reached the market yet. This failure may be attributed to the lack of subtype selectivity for these inhibitors. Therefore, more structural information for subtype sPLA2 is needed to guide the selective inhibitor development. In this study, the crystal structure of human sPLA2 Group IIE (hGIIE), coupled with mutagenesis experiments, proved that the flexible second calcium binding site and residue Asn21 in hGIIE are essential to its enzymatic activity. Five inhibitor bound hGIIE complex structures revealed the key residues (Asn21 and Gly6) of hGIIE that are responsible for interacting with inhibitors, and illustrated the difference in the inhibitor binding pocket with other sPLA2s. This will facilitate the structure-based design of sPLA2's selective inhibitors.
Collapse
Affiliation(s)
- Shulin Hou
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100000, China
| | - Tingting Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Jinxin Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yong Xu
- Guangdong Provincial Key Laboratory of Biocomputing, Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinsong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China. .,Guangdong Provincial Key Laboratory of Biocomputing, Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
26
|
Xia W, Xie L, Cao B, Cheng S, Wan H, Liu H. Genes involved in leukotriene synthesis pathway are dynamically regulated during lung development in Rhesus monkeys. Prostaglandins Leukot Essent Fatty Acids 2017; 122:1-6. [PMID: 28735623 DOI: 10.1016/j.plefa.2017.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/08/2017] [Accepted: 06/13/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND Leukotrienes play critical roles in many inflammatory lung diseases and several antagonists of their receptors have been used in the clinical settings. However, the physiological functions of leukotrienes in lung development are still unclear. METHOD The expression levels of 34 genes involved in leukotriene synthesis and function pathway in the lungs of Rhesus monkey during different developmental time points were determined on a MiSeq platform and analyzed by the reads per kilobase of transcript per million mapped reads (RPKM) method. RESULTS The results showed that the expression levels of PLA2G1B, PLA2G10, PLA2G2D, ALOX5, and ALOX5AP increased dramatically in the lung of Rhesus monkey, reflecting the changes in the pulmonary environment after delivery. Additionally, the different expression patterns between molecules related to LTB4 and LTC4 synthesis suggested distinct roles of LTB4 and LTC4 in lung development. Finally, the constant expression of CysLT1 during the development process provided new information to the pharmaceutical basis of the use of leukotriene receptor antagonists in the clinical setting. CONCLUSION The expression levels of several key genes involved in leukotriene synthesis changed dramatically during lung development in Rhesus monkeys, suggesting the potential roles of leukotrienes in lung development in this animal model.
Collapse
Affiliation(s)
- Wanmin Xia
- Department of Pediatric Respiratory, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Department of Respiratory, Chengdu Women & Children's Hospital, Chengdu 610091, China
| | - Liang Xie
- The Vascular Remodeling and Developmental Defects Research Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Bangrong Cao
- Department of Basic Research, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China; State Key Laboratory of Molecular Oncology, Department of Aetiology and Carcinogenesis, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Aetiology and Carcinogenesis, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Huajing Wan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Lung Development and Diseases, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Hanmin Liu
- Department of Pediatric Respiratory, West China Second University Hospital, Sichuan University, Chengdu 610041, China; The Vascular Remodeling and Developmental Defects Research Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
27
|
Kuefner MS, Pham K, Redd JR, Stephenson EJ, Harvey I, Deng X, Bridges D, Boilard E, Elam MB, Park EA. Secretory phospholipase A 2 group IIA modulates insulin sensitivity and metabolism. J Lipid Res 2017; 58:1822-1833. [PMID: 28663239 DOI: 10.1194/jlr.m076141] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/08/2017] [Indexed: 12/22/2022] Open
Abstract
Secretory phospholipase A2 group IIA (PLA2G2A) is a member of a family of secretory phospholipases that have been implicated in inflammation, atherogenesis, and antibacterial actions. Here, we evaluated the role of PLA2G2A in the metabolic response to a high fat diet. C57BL/6 (BL/6) mice do not express PLA2g2a due to a frameshift mutation. We fed BL/6 mice expressing the human PLA2G2A gene (IIA+ mice) a fat diet and assessed the physiologic response. After 10 weeks on the high fat diet, the BL/6 mice were obese, but the IIA+ mice did not gain weight or accumulate lipid. The lean mass in chow- and high fat-fed IIA+ mice was constant and similar to the BL/6 mice on a chow diet. Surprisingly, the IIA+ mice had an elevated metabolic rate, which was not due to differences in physical activity. The IIA+ mice were more insulin sensitive and glucose tolerant than the BL/6 mice, even when the IIA+ mice were provided the high fat diet. The IIA+ mice had increased expression of uncoupling protein 1 (UCP1), sirtuin 1 (SIRT1), and PPARγ coactivator 1α (PGC-1α) in brown adipose tissue (BAT), suggesting that PLA2G2A activates mitochondrial uncoupling in BAT. Our data indicate that PLA2G2A has a previously undiscovered impact on insulin sensitivity and metabolism.
Collapse
Affiliation(s)
- Michael S Kuefner
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Kevin Pham
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Jeanna R Redd
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Erin J Stephenson
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Innocence Harvey
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Xiong Deng
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Dave Bridges
- Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Faculté de Médecine de l'Université Laval, CHUQ Research Center and Division of Rheumatology, Quebec City, Canada
| | - Marshall B Elam
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN.,Department of Veterans Affairs Medical Center, Memphis, TN
| | - Edwards A Park
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN .,Department of Veterans Affairs Medical Center, Memphis, TN
| |
Collapse
|
28
|
Chen J, Chen L, Sanseau P, Freudenberg JM, Rajpal DK. Significant obesity-associated gene expression changes occur in the stomach but not intestines in obese mice. Physiol Rep 2016; 4:4/10/e12793. [PMID: 27207783 PMCID: PMC4886165 DOI: 10.14814/phy2.12793] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/07/2016] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal (GI) tract can have significant impact on the regulation of the whole‐body metabolism and may contribute to the development of obesity and diabetes. To systemically elucidate the role of the GI tract in obesity, we performed a transcriptomic analysis in different parts of the GI tract of two obese mouse models: ob/ob and high‐fat diet (HFD) fed mice. Compared to their lean controls, significant changes in the gene expression were observed in both obese mouse groups in the stomach (ob/ob: 959; HFD: 542). In addition, these changes were quantitatively much higher than in the intestine. Despite the difference in genetic background, the two mouse models shared 296 similar gene expression changes in the stomach. Among those genes, some had known associations to obesity, diabetes, and insulin resistance. In addition, the gene expression profiles strongly suggested an increased gastric acid secretion in both obese mouse models, probably through an activation of the gastrin pathway. In conclusion, our data reveal a previously unknown dominant connection between the stomach and obesity in murine models extensively used in research.
Collapse
Affiliation(s)
- Jing Chen
- Computational Biology, Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania
| | - Lihong Chen
- Enteroendocrinology DPU, GlaxoSmithKline, Research Triangle Park, North Carolina
| | - Philippe Sanseau
- Computational Biology, Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania
| | | | - Deepak K Rajpal
- Computational Biology, Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania
| |
Collapse
|
29
|
Wang HY, Quan C, Hu C, Xie B, Du Y, Chen L, Yang W, Yang L, Chen Q, Shen B, Hu B, Zheng Z, Zhu H, Huang X, Xu G, Chen S. A lipidomics study reveals hepatic lipid signatures associating with deficiency of the LDL receptor in a rat model. Biol Open 2016; 5:979-86. [PMID: 27378433 PMCID: PMC4958281 DOI: 10.1242/bio.019802] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The low-density lipoprotein receptor (LDLR) plays a critical role in the liver for the clearance of plasma low-density lipoprotein (LDL). Its deficiency causes hypercholesterolemia in many models. To facilitate the usage of rats as animal models for the discovery of cholesterol-lowering drugs, we took a genetic approach to delete the LDLR in rats aiming to increase plasma LDL cholesterol (LDL-C). An LDLR knockout rat was generated via zinc-finger nuclease technology, which harbors a 19-basepair deletion in the seventh exon of the ldlr gene. As expected, deletion of the LDLR elevated total cholesterol and total triglyceride in the plasma, and caused a tenfold increase of plasma LDL-C and a fourfold increase of plasma very low-density lipoprotein (VLDL-C). A lipidomics analysis revealed that deletion of the LDLR affected hepatic lipid metabolism, particularly lysophosphatidylcholines, free fatty acids and sphingolipids in the liver. Cholesterol ester (CE) 20:4 also displayed a significant increase in the LDLR knockout rats. Taken together, the LDLR knockout rat offers a new model of hypercholesterolemia, and the lipidomics analysis reveals hepatic lipid signatures associating with deficiency of the LDL receptor. Summary: An LDL receptor knockout rat model was generated which offers a new hypercholesterolemia model. A lipidomics analysis reveals hepatic lipid signatures associating with LDLR deficiency in rats.
Collapse
Affiliation(s)
- Hong Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China
| | - Chao Quan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Yinan Du
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Liang Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Wei Yang
- Laboratory Animal Center, China Medical University, Shenyang 110001, China
| | - Liu Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Bin Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Bian Hu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Zhihong Zheng
- Laboratory Animal Center, China Medical University, Shenyang 110001, China
| | - Haibo Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xingxu Huang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China
| |
Collapse
|
30
|
Sato H, Taketomi Y, Murakami M. Metabolic regulation by secreted phospholipase A 2. Inflamm Regen 2016; 36:7. [PMID: 29259680 PMCID: PMC5725825 DOI: 10.1186/s41232-016-0012-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/10/2016] [Indexed: 12/18/2022] Open
Abstract
Within the phospholipase A2 (PLA2) superfamily that hydrolyzes phospholipids to yield fatty acids and lysophospholipids, the secreted PLA2 (sPLA2) enzymes comprise the largest family that contains 11 isoforms in mammals. Individual sPLA2s exhibit unique distributions and specific enzymatic properties, suggesting their distinct biological roles. While sPLA2s have long been implicated in inflammation and atherosclerosis, it has become evident that they are involved in diverse biological events through lipid mediator-dependent or mediator-independent processes in a given microenvironment. In recent years, new biological aspects of sPLA2s have been revealed using their transgenic and knockout mouse models in combination with mass spectrometric lipidomics to unveil their target substrates and products in vivo. In this review, we summarize our current knowledge of the roles of sPLA2s in metabolic disorders including obesity, hepatic steatosis, diabetes, insulin resistance, and adipose tissue inflammation.
Collapse
Affiliation(s)
- Hiroyasu Sato
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Yoshitaka Taketomi
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Makoto Murakami
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, 100-0004 Japan
| |
Collapse
|
31
|
Liver-specific overexpression of LPCAT3 reduces postprandial hyperglycemia and improves lipoprotein metabolic profile in mice. Nutr Diabetes 2016; 6:e206. [PMID: 27110687 PMCID: PMC4855257 DOI: 10.1038/nutd.2016.12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/12/2016] [Accepted: 03/14/2016] [Indexed: 01/09/2023] Open
Abstract
Previous studies have shown that group 1B phospholipase A2-mediated absorption of lysophospholipids inhibits hepatic fatty acid β-oxidation and contributes directly to postprandial hyperglycemia and hyperlipidemia, leading to increased risk of cardiometabolic disease. The current study tested the possibility that increased expression of lysophosphatidylcholine acyltransferase-3 (LPCAT3), an enzyme that converts lysophosphatidylcholine to phosphatidylcholine in the liver, may alleviate the adverse effects of lysophospholipids absorbed after a lipid-glucose mixed meal. The injection of an adenovirus vector harboring the human LPCAT3 gene into C57BL/6 mice increased hepatic LPCAT3 expression fivefold compared with mice injected with a control LacZ adenovirus. Postprandial glucose tolerance tests after feeding these animals with a bolus lipid-glucose mixed meal revealed that LPCAT3 overexpression improved postprandial hyperglycemia and glucose tolerance compared with control mice with LacZ adenovirus injection. Mice with LPCAT3 overexpression also showed reduced very low density lipoprotein production and displayed elevated levels of the metabolic- and cardiovascular-protective large apoE-rich high density lipoproteins in plasma. The mechanism underlying the metabolic benefits of LPCAT3 overexpression was shown to be due to the alleviation of lysophospholipid inhibition of fatty acid β-oxidation in hepatocytes. Taken together, these results suggest that specific LPCAT3 induction in the liver may be a viable strategy for cardiometabolic disease intervention.
Collapse
|
32
|
Chap H. Forty five years with membrane phospholipids, phospholipases and lipid mediators: A historical perspective. Biochimie 2016; 125:234-49. [PMID: 27059515 DOI: 10.1016/j.biochi.2016.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 01/02/2023]
Abstract
Phospholipases play a key role in the metabolism of phospholipids and in cell signaling. They are also a very useful tool to explore phospholipid structure and metabolism as well as membrane organization. They are at the center of this review, covering a period starting in 1971 and focused on a number of subjects in which my colleagues and I have been involved. Those include determination of phospholipid asymmetry in the blood platelet membrane, biosynthesis of lysophosphatidic acid, biochemistry of platelet-activating factor, first attempts to define the role of phosphoinositides in cell signaling, and identification of novel digestive (phospho)lipases such as pancreatic lipase-related protein 2 (PLRP2) or phospholipase B. Besides recalling some of our contributions to those various fields, this review makes an appraisal of the impressive and often unexpected evolution of those various aspects of membrane phospholipids and lipid mediators. It is also the occasion to propose some new working hypotheses.
Collapse
Affiliation(s)
- Hugues Chap
- Centre de Physiopathologie de Toulouse Purpan, Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France. hugues.chap.@univ-tlse3.fr
| |
Collapse
|
33
|
Wang M, Wang C, Han RH, Han X. Novel advances in shotgun lipidomics for biology and medicine. Prog Lipid Res 2016; 61:83-108. [PMID: 26703190 PMCID: PMC4733395 DOI: 10.1016/j.plipres.2015.12.002] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
The field of lipidomics, as coined in 2003, has made profound advances and been rapidly expanded. The mass spectrometry-based strategies of this analytical methodology-oriented research discipline for lipid analysis are largely fallen into three categories: direct infusion-based shotgun lipidomics, liquid chromatography-mass spectrometry-based platforms, and matrix-assisted laser desorption/ionization mass spectrometry-based approaches (particularly in imagining lipid distribution in tissues or cells). This review focuses on shotgun lipidomics. After briefly introducing its fundamentals, the major materials of this article cover its recent advances. These include the novel methods of lipid extraction, novel shotgun lipidomics strategies for identification and quantification of previously hardly accessible lipid classes and molecular species including isomers, and novel tools for processing and interpretation of lipidomics data. Representative applications of advanced shotgun lipidomics for biological and biomedical research are also presented in this review. We believe that with these novel advances in shotgun lipidomics, this approach for lipid analysis should become more comprehensive and high throughput, thereby greatly accelerating the lipidomics field to substantiate the aberrant lipid metabolism, signaling, trafficking, and homeostasis under pathological conditions and their underpinning biochemical mechanisms.
Collapse
Affiliation(s)
- Miao Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Rowland H Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA; College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
34
|
Wang C, Wang M, Han X. Comprehensive and quantitative analysis of lysophospholipid molecular species present in obese mouse liver by shotgun lipidomics. Anal Chem 2015; 87:4879-87. [PMID: 25860968 DOI: 10.1021/acs.analchem.5b00410] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Shotgun lipidomics exploits the unique chemical and physical properties of lipid classes and individual molecular species to facilitate the high-throughput analysis of a cellular lipidome on a large scale directly from the extracts of biological samples. A platform for comprehensive analysis of lysophospholipid (LPL) species based on shotgun lipidomics has not been established. Herein, after extensive characterization of the fragmentation pattern of individual LPL class and optimization of all experimental conditions including developing new methods for optimization of collision energy, and recovery and enrichment of LPL classes from the aqueous phase after solvent extraction, a new method for comprehensive and quantitative analysis of LPL species was developed. This newly developed method was applied for comprehensive analysis of LPL species present in mouse liver samples. Remarkably, the study revealed significant accumulation of LPL species in the liver of ob/ob mice. Taken together, by exploiting the principles of shotgun lipidomics in combination with a novel strategy of sample preparation, LPL species present in biological samples can be determined by the established method. We believe that this development is significant and useful for understanding the pathways of phospholipid metabolism and for elucidating the role of LPL species in signal transduction and other biological functions.
Collapse
Affiliation(s)
- Chunyan Wang
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, United States
| | - Miao Wang
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, United States
| | - Xianlin Han
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, United States
| |
Collapse
|
35
|
Murakami M, Sato H, Miki Y, Yamamoto K, Taketomi Y. A new era of secreted phospholipase A₂. J Lipid Res 2015; 56:1248-61. [PMID: 25805806 DOI: 10.1194/jlr.r058123] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Indexed: 12/18/2022] Open
Abstract
Among more than 30 members of the phospholipase A2 (PLA2) superfamily, secreted PLA2 (sPLA2) enzymes represent the largest family, being Ca(2+)-dependent low-molecular-weight enzymes with a His-Asp catalytic dyad. Individual sPLA2s exhibit unique tissue and cellular distributions and enzymatic properties, suggesting their distinct biological roles. Recent studies using transgenic and knockout mice for nearly a full set of sPLA2 subtypes, in combination with sophisticated lipidomics as well as biochemical and cell biological studies, have revealed distinct contributions of individual sPLA2s to various pathophysiological events, including production of pro- and anti-inflammatory lipid mediators, regulation of membrane remodeling, degradation of foreign phospholipids in microbes or food, or modification of extracellular noncellular lipid components. In this review, we highlight the current understanding of the in vivo functions of sPLA2s and the underlying lipid pathways as revealed by a series of studies over the last decade.
Collapse
Affiliation(s)
- Makoto Murakami
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan CREST, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Hiroyasu Sato
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yoshimi Miki
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kei Yamamoto
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yoshitaka Taketomi
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
36
|
Navab M, Chattopadhyay A, Hough G, Meriwether D, Fogelman SI, Wagner AC, Grijalva V, Su F, Anantharamaiah GM, Hwang LH, Faull KF, Reddy ST, Fogelman AM. Source and role of intestinally derived lysophosphatidic acid in dyslipidemia and atherosclerosis. J Lipid Res 2015; 56:871-87. [PMID: 25646365 DOI: 10.1194/jlr.m056614] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We previously reported that i) a Western diet increased levels of unsaturated lysophosphatidic acid (LPA) in small intestine and plasma of LDL receptor null (LDLR(-/-)) mice, and ii) supplementing standard mouse chow with unsaturated (but not saturated) LPA produced dyslipidemia and inflammation. Here we report that supplementing chow with unsaturated (but not saturated) LPA resulted in aortic atherosclerosis, which was ameliorated by adding transgenic 6F tomatoes. Supplementing chow with lysophosphatidylcholine (LysoPC) 18:1 (but not LysoPC 18:0) resulted in dyslipidemia similar to that seen on adding LPA 18:1 to chow. PF8380 (a specific inhibitor of autotaxin) significantly ameliorated the LysoPC 18:1-induced dyslipidemia. Supplementing chow with LysoPC 18:1 dramatically increased the levels of unsaturated LPA species in small intestine, liver, and plasma, and the increase was significantly ameliorated by PF8380 indicating that the conversion of LysoPC 18:1 to LPA 18:1 was autotaxin dependent. Adding LysoPC 18:0 to chow increased levels of LPA 18:0 in small intestine, liver, and plasma but was not altered by PF8380 indicating that conversion of LysoPC 18:0 to LPA 18:0 was autotaxin independent. We conclude that i) intestinally derived unsaturated (but not saturated) LPA can cause atherosclerosis in LDLR(-/-) mice, and ii) autotaxin mediates the conversion of unsaturated (but not saturated) LysoPC to LPA.
Collapse
Affiliation(s)
- Mohamad Navab
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Arnab Chattopadhyay
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Greg Hough
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - David Meriwether
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Spencer I Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan C Wagner
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Victor Grijalva
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Feng Su
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lin H Hwang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Srinivasa T Reddy
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| |
Collapse
|
37
|
Murakami M, Taketomi Y, Miki Y, Sato H, Yamamoto K, Lambeau G. Emerging roles of secreted phospholipase A2 enzymes: the 3rd edition. Biochimie 2014; 107 Pt A:105-13. [PMID: 25230085 DOI: 10.1016/j.biochi.2014.09.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/05/2014] [Indexed: 12/19/2022]
Abstract
Within the phospholipase A2 (PLA2) superfamily, secreted PLA2 (sPLA2) enzymes comprise the largest family that contains 11 to 12 mammalian isoforms with a conserved His-Asp catalytic dyad. Individual sPLA2s exhibit unique tissue and cellular localizations and specific enzymatic properties, suggesting distinct biological roles. Individual sPLA2s are involved in diverse biological events through lipid mediator-dependent or -independent processes and act redundantly or non-redundantly in a given microenvironment. In the past few years, new biological aspects of sPLA2s have been clarified using their transgenic and knockout mouse lines in combination with mass spectrometric lipidomics to unveil their target substrates and products in vivo. In the 3rd edition of this review series, we highlight the newest understanding of the in vivo functions of sPLA2s in pathophysiological conditions in the context of immunity and metabolism. We will also describe the latest knowledge on PLA2R1, the best known sPLA2 receptor, which may serve either as a clearance or signaling receptor for sPLA2 or may even act independently of sPLA2 function.
Collapse
Affiliation(s)
- Makoto Murakami
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; CREST, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| | - Yoshitaka Taketomi
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Yoshimi Miki
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Hiroyasu Sato
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kei Yamamoto
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Gérard Lambeau
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Centre National de la Recherche Scientifique - Université Nice Sophia Antipolis, Valbonne 06560, France
| |
Collapse
|
38
|
Hollie NI, Konaniah ES, Goodin C, Hui DY. Group 1B phospholipase A₂ inactivation suppresses atherosclerosis and metabolic diseases in LDL receptor-deficient mice. Atherosclerosis 2014; 234:377-80. [PMID: 24747111 PMCID: PMC4037866 DOI: 10.1016/j.atherosclerosis.2014.03.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Previous studies have shown that inactivation of the group 1B phospholipase A2 (Pla2g1b) suppresses diet-induced obesity, hyperglycemia, insulin resistance, and hyperlipidemia in C57BL/6 mice. A possible influence of Pla2g1b inactivation on atherosclerosis has not been addressed previously. The current study utilized LDL receptor-deficient (Ldlr(-/-)) mice with plasma lipid levels and distribution similar to hyperlipidemic human subjects as a preclinical animal model to test the effectiveness of Pla2g1b inactivation on atherosclerosis. METHODS AND RESULTS The Pla2g1b(+/+)Ldlr(-/-) and Pla2g1b(-/-)Ldlr(-/-) mice were fed a low fat chow diet or a hypercaloric diet with 58.5 kcal% fat and 25 kcal% sucrose for 10 weeks. Minimal differences were observed between Pla2g1b(+/+)Ldlr(-/-) and Pla2g1b(-/-)Ldlr(-/-) mice when the animals were maintained on the low fat chow diet. However, when the animals were maintained on the hypercaloric diet, the Pla2g1(+/+)Ldlr(-/-) mice showed the expected body weight gain but the Pla2g1b(-/-)Ldlr(-/-) mice were resistant to diet-induced body weight gain. The Pla2g1b(-/-)Ldlr(-/-) mice also displayed lower fasting glucose, insulin, and plasma lipid levels compared to the Pla2g1b(+/+)Ldlr(-/-) mice, which displayed robust hyperglycemia, hyperinsulinemia, and hyperlipidemia in response to the hypercaloric diet. Importantly, atherosclerotic lesions in the aortic roots were also reduced 7-fold in the Pla2g1b(-/-)Ldlr(-/-) mice. CONCLUSION The effectiveness of Pla2g1b inactivation to suppress diet-induced body weight gain and reduce diabetes and atherosclerosis in LDL receptor-deficient mice suggests that pharmacological inhibition of Pla2g1b may be a viable strategy to decrease diet-induced obesity and the risk of diabetes and atherosclerosis in humans.
Collapse
Affiliation(s)
- Norris I Hollie
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Eddy S Konaniah
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - Colleen Goodin
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA.
| |
Collapse
|
39
|
Hollie NI, Cash JG, Matlib MA, Wortman M, Basford JE, Abplanalp W, Hui DY. Micromolar changes in lysophosphatidylcholine concentration cause minor effects on mitochondrial permeability but major alterations in function. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:888-95. [PMID: 24315825 DOI: 10.1016/j.bbalip.2013.11.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/02/2013] [Accepted: 11/26/2013] [Indexed: 12/17/2022]
Abstract
Mice deficient in group 1b phospholipase A2 have decreased plasma lysophosphatidylcholine and increased hepatic oxidation that is inhibited by intraperitoneal lysophosphatidylcholine injection. This study sought to identify a mechanism for lysophosphatidylcholine-mediated inhibition of hepatic oxidative function. Results showed that in vitro incubation of isolated mitochondria with 40-200μM lysophosphatidylcholine caused cyclosporine A-resistant swelling in a concentration-dependent manner. However, when mitochondria were challenged with 220μM CaCl2, cyclosporine A protected against permeability transition induced by 40μM, but not 80μM lysophosphatidylcholine. Incubation with 40-120μM lysophosphatidylcholine also increased mitochondrial permeability to 75μM CaCl2 in a concentration-dependent manner. Interestingly, despite incubation with 80μM lysophosphatidylcholine, the mitochondrial membrane potential was steady in the presence of succinate, and oxidation rates and respiratory control indices were similar to controls in the presence of succinate, glutamate/malate, and palmitoyl-carnitine. However, mitochondrial oxidation rates were inhibited by 30-50% at 100μM lysophosphatidylcholine. Finally, while 40μM lysophosphatidylcholine has no effect on fatty acid oxidation and mitochondria remained impermeable in intact hepatocytes, 100μM lysophosphatidylcholine inhibited fatty acid stimulated oxidation and caused intracellular mitochondrial permeability. Taken together, these present data demonstrated that LPC concentration dependently modulates mitochondrial microenvironment, with low micromolar concentrations of lysophosphatidylcholine sufficient to change hepatic oxidation rate whereas higher concentrations are required to disrupt mitochondrial integrity.
Collapse
Affiliation(s)
- Norris I Hollie
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James G Cash
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M Abdul Matlib
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew Wortman
- Department of Internal Medicine, Division of Endocrinology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joshua E Basford
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William Abplanalp
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
40
|
Wu HT, Chen W, Cheng KC, Ku PM, Yeh CH, Cheng JT. Oleic acid activates peroxisome proliferator-activated receptor δ to compensate insulin resistance in steatotic cells. J Nutr Biochem 2012; 23:1264-70. [DOI: 10.1016/j.jnutbio.2011.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 07/09/2011] [Accepted: 07/14/2011] [Indexed: 01/22/2023]
|
41
|
Abstract
Non-systemic drugs act within the intestinal lumen without reaching the systemic circulation. The first generation included polymeric resins that sequester phosphate ions, potassium ions, or bile acids for the treatment of electrolyte imbalances or hypercholesteremia. The field has evolved towards non-absorbable small molecules or peptides targeting luminal enzymes or transporters for the treatment of mineral metabolism disorders, diabetes, gastrointestinal (GI) disorders, and enteric infections. From a drug design and development perspective, non-systemic agents offer novel opportunities to address unmet medical needs while minimizing toxicity risks, but also present new challenges, including developing a better understanding and control of non-transcellular leakage pathways into the systemic circulation. The pharmacokinetic-pharmacodynamic relationship of drugs acting in the GI tract can be complex due to the variability of intestinal transit, interaction with chyme, and the complex environment of the surface epithelia. We review the main classes of nonabsorbable agents at various stages of development, and their therapeutic potential and limitations. The rapid progress in the identification of intestinal receptors and transporters, their functional characterization and role in metabolic and inflammatory disorders, will undoubtedly renew interest in the development of novel, safe, non-systemic therapeutics.
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The phospholipase A2 (PLA2) family of proteins includes lipolytic enzymes that liberate the sn-2 fatty acyl chains from phospholipids to yield nonesterified fatty acids and lysophospholipids. The purpose of this review is to discuss recent findings showing distinct roles of several of these PLA2 enzymes in inflammatory metabolic diseases such as diabetes and atherosclerosis. RECENT FINDINGS The group 1B PLA2 digestion of phospholipids in the intestinal lumen facilitates postprandial lysophospholipid absorption, which suppresses hepatic fatty acid oxidation leading to increased VLDL synthesis, decreased glucose tolerance, and promotion of tissue lipid deposition to accentuate diet-induced hyperlipidemia, diabetes, and obesity. Other secretory PLA2s promote inflammatory metabolic diseases by generating bioactive lipid metabolites to induce inflammatory cytokine production, whereas the major intracellular PLA2s, cPLA2α, and iPLA2, generate arachidonic acid and lysophosphatic acid in response to extracellular stimuli to activate leukocyte chemotactic response. SUMMARY Each member of the PLA2 family of enzymes serves a distinct role in generating active lipid metabolites that promote inflammatory metabolic diseases including atherosclerosis, hyperlipidemia, obesity, and diabetes. The development of specific drugs that target one or more of these PLA2 enzymes may be novel strategies for treatment of these chronic inflammatory metabolic disorders.
Collapse
Affiliation(s)
- David Y Hui
- Department of Pathology, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
43
|
Hollie NI, Hui DY. Group 1B phospholipase A₂ deficiency protects against diet-induced hyperlipidemia in mice. J Lipid Res 2011; 52:2005-11. [PMID: 21908646 DOI: 10.1194/jlr.m019463] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Excessive absorption of products of dietary fat digestion leads to type 2 diabetes and other obesity-related disorders. Mice deficient in the group 1B phospholipase A₂ (Pla2g1b), a gut digestive enzyme, are protected against diet-induced obesity and type 2 diabetes without displaying dietary lipid malabsorption. This study tested the hypothesis that inhibition of Pla2g1b protects against diet-induced hyperlipidemia. Results showed that the Pla2g1b(-/-) mice had decreased plasma triglyceride and cholesterol levels compared with Pla2g1b(+/+) mice subsequent to feeding a high-fat, high-carbohydrate (hypercaloric) diet. These differences were evident before differences in body weight gains were observed. Injection of Poloxamer 407 to inhibit lipolysis revealed decreased VLDL production in Pla2g1b(-/-) mice. Supplementation with lysophosphatidylcholine, the product of Pla2g1b hydrolysis, restored VLDL production rates in Pla2g1b(-/-) mice and further elevated VLDL production in Pla2g1b(+/+) mice. The Pla2g1b(-/-) mice also displayed decreased postprandial lipidemia compared with Pla2g1b(+/+) mice. These results show that, in addition to dietary fatty acids, gut-derived lysophospholipids derived from Pla2g1b hydrolysis of dietary and biliary phospholipids also promote hepatic VLDL production. Thus, the inhibition of lysophospholipid absorption via Pla2g1b inactivation may prove beneficial against diet-induced hyperlipidemia in addition to the protection against obesity and diabetes.
Collapse
Affiliation(s)
- Norris I Hollie
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | | |
Collapse
|
44
|
Sato H, Isogai Y, Masuda S, Taketomi Y, Miki Y, Kamei D, Hara S, Kobayashi T, Ishikawa Y, Ishii T, Ikeda K, Taguchi R, Ishimoto Y, Suzuki N, Yokota Y, Hanasaki K, Suzuki-Yamamoto T, Yamamoto K, Murakami M. Physiological roles of group X-secreted phospholipase A2 in reproduction, gastrointestinal phospholipid digestion, and neuronal function. J Biol Chem 2011; 286:11632-48. [PMID: 21266581 PMCID: PMC3064216 DOI: 10.1074/jbc.m110.206755] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 01/17/2011] [Indexed: 01/04/2023] Open
Abstract
Although the secreted phospholipase A(2) (sPLA(2)) family has been generally thought to participate in pathologic events such as inflammation and atherosclerosis, relatively high and constitutive expression of group X sPLA(2) (sPLA(2)-X) in restricted sites such as reproductive organs, the gastrointestinal tract, and peripheral neurons raises a question as to the roles played by this enzyme in the physiology of reproduction, digestion, and the nervous system. Herein we used mice with gene disruption or transgenic overexpression of sPLA(2)-X to clarify the homeostatic functions of this enzyme at these locations. Our results suggest that sPLA(2)-X regulates 1) the fertility of spermatozoa, not oocytes, beyond the step of flagellar motility, 2) gastrointestinal phospholipid digestion, perturbation of which is eventually linked to delayed onset of a lean phenotype with reduced adiposity, decreased plasma leptin, and improved muscle insulin tolerance, and 3) neuritogenesis of dorsal root ganglia and the duration of peripheral pain nociception. Thus, besides its inflammatory action proposed previously, sPLA(2)-X participates in physiologic processes including male fertility, gastrointestinal phospholipid digestion linked to adiposity, and neuronal outgrowth and sensing.
Collapse
Affiliation(s)
- Hiroyasu Sato
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Yuki Isogai
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- the Department of Biology, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610
| | - Seiko Masuda
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Yoshitaka Taketomi
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Yoshimi Miki
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Daisuke Kamei
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Shuntaro Hara
- the Department of Health Chemistry, School of Pharceutical Sciences, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555
| | - Tetsuyuki Kobayashi
- the Department of Biology, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610
| | - Yukio Ishikawa
- the Department of Pathology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ohta-ku, Tokyo 143-8540
| | - Toshiharu Ishii
- the Department of Pathology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ohta-ku, Tokyo 143-8540
| | - Kazutaka Ikeda
- the Department of Metabolome, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033
- the Department of Neutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Kuboki 111, Souja, Okayama 719-1197, and
| | - Ryo Taguchi
- the Department of Metabolome, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033
- CREST and
| | - Yoshikazu Ishimoto
- Shionogi Research Laboratories, Shionogi and Company Ltd, 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825
| | - Noriko Suzuki
- Shionogi Research Laboratories, Shionogi and Company Ltd, 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825
| | - Yasunori Yokota
- Shionogi Research Laboratories, Shionogi and Company Ltd, 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825
| | - Kohji Hanasaki
- Shionogi Research Laboratories, Shionogi and Company Ltd, 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825
| | - Toshiko Suzuki-Yamamoto
- the Department of Neutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Kuboki 111, Souja, Okayama 719-1197, and
| | - Kei Yamamoto
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
| | - Makoto Murakami
- From the Lipid Metabolism Project, the Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 256-8506
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
45
|
Murakami M, Taketomi Y, Miki Y, Sato H, Hirabayashi T, Yamamoto K. Recent progress in phospholipase A₂ research: from cells to animals to humans. Prog Lipid Res 2010; 50:152-92. [PMID: 21185866 DOI: 10.1016/j.plipres.2010.12.001] [Citation(s) in RCA: 368] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mammalian genomes encode genes for more than 30 phospholipase A₂s (PLA₂s) or related enzymes, which are subdivided into several classes including low-molecular-weight secreted PLA₂s (sPLA₂s), Ca²+-dependent cytosolic PLA₂s (cPLA₂s), Ca²+-independent PLA₂s (iPLA₂s), platelet-activating factor acetylhydrolases (PAF-AHs), lysosomal PLA₂s, and a recently identified adipose-specific PLA. Of these, the intracellular cPLA₂ and iPLA₂ families and the extracellular sPLA₂ family are recognized as the "big three". From a general viewpoint, cPLA₂α (the prototypic cPLA₂ plays a major role in the initiation of arachidonic acid metabolism, the iPLA₂ family contributes to membrane homeostasis and energy metabolism, and the sPLA₂ family affects various biological events by modulating the extracellular phospholipid milieus. The cPLA₂ family evolved along with eicosanoid receptors when vertebrates first appeared, whereas the diverse branching of the iPLA₂ and sPLA₂ families during earlier eukaryote development suggests that they play fundamental roles in life-related processes. During the past decade, data concerning the unexplored roles of various PLA₂ enzymes in pathophysiology have emerged on the basis of studies using knockout and transgenic mice, the use of specific inhibitors, and information obtained from analysis of human diseases caused by mutations in PLA₂ genes. This review focuses on current understanding of the emerging biological functions of PLA₂s and related enzymes.
Collapse
Affiliation(s)
- Makoto Murakami
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Pancreatic acinar cell-specific overexpression of group 1B phospholipase A2 exacerbates diet-induced obesity and insulin resistance in mice. Int J Obes (Lond) 2010; 35:877-81. [PMID: 20938441 DOI: 10.1038/ijo.2010.215] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Genome-wide association studies have identified significant association between polymorphisms of the Group 1B phospholipase A(2) (PLA2G1B) gene and central obesity in humans. Previous studies have shown that Pla2g1b inactivation decreases post-prandial lysophospholipid absorption, and as a consequence increases hepatic fatty acid oxidation and protects against diet-induced obesity and glucose intolerance in mice. The present study showed that transgenic mice with pancreatic acinar cell-specific overexpression of the human PLA2G1B gene gained significantly more weight and displayed elevated insulin resistance characteristics, such as impaired glucose tolerance, compared with wild-type (WT) mice, when challenged with a high-fat/carbohydrate diet. Pre- and post-prandial plasma β-hydroxybutyrate levels were also lower, indicative of decreased hepatic fatty acid oxidation, in the hypercaloric diet-fed PLA2G1B transgenic mice. These, along with earlier observations of Pla2g1b-null mice, document that Pla2g1b expression level is an important determinant of susceptibility to diet-induced obesity and diabetes, suggesting that the relationship between PLA2G1B polymorphisms and obesity may be due to differences in PLA2G1B expression levels between these individuals. The ability of pancreas-specific overexpression of PLA2G1B to promote obesity and glucose intolerance suggests that target phospholipase activity in the digestive tract with non-absorbable inhibitors should be considered as a therapeutic option for metabolic disease therapy.
Collapse
|
47
|
Li X, Shridas P, Forrest K, Bailey W, Webb NR. Group X secretory phospholipase A2 negatively regulates adipogenesis in murine models. FASEB J 2010; 24:4313-24. [PMID: 20585029 DOI: 10.1096/fj.10-154716] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Studies in vitro indicate that group X secretory phospholipase A(2) (GX sPLA(2)) potently releases arachidonic acid (AA) and lysophosphatidylcholine from mammalian cell membranes. To define the function of GX sPLA(2) in vivo, our laboratory recently generated C57BL/6 mice with targeted deletion of GX sPLA(2) (GX(-/-) mice). When fed a normal rodent diet, GX(-/-) mice gained significantly more weight and had increased adiposity compared to GX(+/+) mice, which was not attributable to alterations in food consumption or energy expenditure. When treated with adipogenic stimuli ex vivo, stromal vascular cells isolated from adipose tissue of GX(-/-) mice accumulated significantly more (20%) triglyceride compared to cells from GX(+/+) mice. Conversely, overexpression of GX sPLA(2), but not catalytically inactive GX sPLA(2), resulted in a significant 50% reduction in triglyceride accumulation in OP9 adipocytes. The induction of genes encoding adipogenic proteins (PPARγ, SREBP-1c, SCD1, and FAS) was also significantly blunted by 50-80% in OP9 cells overexpressing GX sPLA(2). Activation of the liver X receptor (LXR), a nuclear receptor known to up-regulate adipogenic gene expression, was suppressed in 3T3-L1 and OP9 cells when GX sPLA(2) was overexpressed. Thus, hydrolytic products generated by GX sPLA(2) negatively regulate adipogenesis, possibly by suppressing LXR activation.
Collapse
Affiliation(s)
- Xia Li
- Graduate Center for Nutritional Sciences, University of Kentucky Medical Center, Lexington, KY 40536-0200, USA
| | | | | | | | | |
Collapse
|