1
|
Hara A, Lu E, Johnstone L, Wei M, Sun S, Hallmark B, Watkins JC, Zhang HH, Yao G, Chilton FH. Identification of an Allele-Specific Transcription Factor Binding Interaction that May Regulate PLA2G2A Gene Expression. Bioinform Biol Insights 2024; 18:11779322241261427. [PMID: 39081667 PMCID: PMC11287738 DOI: 10.1177/11779322241261427] [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: 02/29/2024] [Accepted: 05/24/2024] [Indexed: 08/02/2024] Open
Abstract
The secreted phospholipase A2 (sPLA2) isoform, sPLA2-IIA, has been implicated in a variety of diseases and conditions, including bacteremia, cardiovascular disease, COVID-19, sepsis, adult respiratory distress syndrome, and certain cancers. Given its significant role in these conditions, understanding the regulatory mechanisms impacting its levels is crucial. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs), including rs11573156, that are associated with circulating levels of sPLA2-IIA. The work in the manuscript leveraged 4 publicly available datasets to investigate the mechanism by which rs11573156 influences sPLA2-IIA levels via bioinformatics and modeling analysis. Through genotype-tissue expression (GTEx), 234 expression quantitative trait loci (eQTLs) were identified for the gene that encodes for sPLA2-IIA, PLA2G2A. SNP2TFBS was used to ascertain the binding affinities between transcription factors (TFs) to both the reference and alternative alleles of identified eQTL SNPs. Subsequently, candidate TF-SNP interactions were cross-referenced with the ChIP-seq results in matched tissues from ENCODE. SP1-rs11573156 emerged as the significant TF-SNP pair in the liver. Further analysis revealed that the upregulation of PLA2G2A transcript levels through the rs11573156 variant was likely affected by tissue SP1 protein levels. Using an ordinary differential equation based on Michaelis-Menten kinetic assumptions, we modeled the dependence of PLA2G2A transcription on SP1 protein levels, incorporating the SNP influence. Collectively, our analysis strongly suggests that the difference in the binding dynamics of SP1 to different rs11573156 alleles may underlie the allele-specific PLA2G2A expression in different tissues, a mechanistic model that awaits future direct experimental validation. This mechanism likely contributes to the variation in circulating sPLA2-IIA protein levels in the human population, with implications for a wide range of human diseases.
Collapse
Affiliation(s)
- Aki Hara
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
| | - Eric Lu
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Laurel Johnstone
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
| | - Michelle Wei
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Shudong Sun
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Brian Hallmark
- BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - Joseph C Watkins
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Hao Helen Zhang
- Department of Mathematics, The University of Arizona, Tucson, AZ, USA
- Statistics Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA
| | - Guang Yao
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, USA
- Center for Precision Nutrition and Wellness, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
2
|
Hara A, Lu E, Johnstone L, Wei M, Sun S, Hallmark B, Watkins JC, Zhang HH, Yao G, Chilton FH. Identification of an allele-specific transcription factor binding interaction that regulates PLA2G2A gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571290. [PMID: 38168258 PMCID: PMC10760018 DOI: 10.1101/2023.12.12.571290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The secreted phospholipase A 2 (sPLA 2 ) isoform, sPLA 2 -IIA, has been implicated in a variety of diseases and conditions, including bacteremia, cardiovascular disease, COVID-19, sepsis, adult respiratory distress syndrome, and certain cancers. Given its significant role in these conditions, understanding the regulatory mechanisms impacting its levels is crucial. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs), including rs11573156, that are associated with circulating levels of sPLA 2 -IIA. Through Genotype-Tissue Expression (GTEx), 234 expression quantitative trait loci (eQTLs) were identified for the gene that encodes for sPLA 2 -IIA, PLA2G2A . SNP2TFBS ( https://ccg.epfl.ch/snp2tfbs/ ) was utilized to ascertain the binding affinities between transcription factors (TFs) to both the reference and alternative alleles of identified SNPs. Subsequently, ChIP-seq peaks highlighted the TF combinations that specifically bind to the SNP, rs11573156. SP1 emerged as a significant TF/SNP pair in liver cells, with rs11573156/SP1 interaction being most prominent in liver, prostate, ovary, and adipose tissues. Further analysis revealed that the upregulation of PLA2G2A transcript levels through the rs11573156 variant was affected by tissue SP1 protein levels. By leveraging an ordinary differential equation, structured upon Michaelis-Menten enzyme kinetics assumptions, we modeled the PLA2G2A transcription's dependence on SP1 protein levels, incorporating the SNP's influence. Collectively, these data strongly suggest that the binding affinity differences of SP1 for the different rs11573156 alleles can influence PLA2G2A expression. This, in turn, can modulate sPLA2-IIA levels, impacting a wide range of human diseases.
Collapse
|
3
|
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: 27] [Impact Index Per Article: 13.5] [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
|
4
|
Yamamoto K, Hakoi H, Nomura S, Murakami M. The Roles of sPLA 2s in Skin Homeostasis and Disease. Biomolecules 2023; 13:biom13040668. [PMID: 37189415 DOI: 10.3390/biom13040668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/21/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Among the phospholipase A2 (PLA2) family, the secreted PLA2 (sPLA2) family in mammals contains 11 members that exhibit unique tissue or cellular distributions and enzymatic properties. Current studies using knockout and/or transgenic mice for a nearly full set of sPLA2s, in combination with comprehensive lipidomics, have revealed the diverse pathophysiological roles of sPLA2s in various biological events. Individual sPLA2s exert specific functions within tissue microenvironments, likely through the hydrolysis of extracellular phospholipids. Lipids are an essential biological component for skin homeostasis, and disturbance of lipid metabolism by deletion or overexpression of lipid-metabolizing enzymes or lipid-sensing receptors often leads to skin abnormalities that are easily visible on the outside. Over the past decades, our studies using knockout and transgenic mice for various sPLA2s have uncovered several new aspects of these enzymes as modulators of skin homeostasis and disease. This article summarizes the roles of several sPLA2s in skin pathophysiology, providing additional insight into the research fields of sPLA2s, lipids, and skin biology.
Collapse
Affiliation(s)
- Kei Yamamoto
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minami-jyosanjima, Tokushima 770-8513, Japan
| | - Haruka Hakoi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minami-jyosanjima, Tokushima 770-8513, Japan
| | - Saki Nomura
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minami-jyosanjima, Tokushima 770-8513, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo (UTokyo), 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
5
|
Secretory Phospholipases A2, from Snakebite Envenoming to a Myriad of Inflammation Associated Human Diseases-What Is the Secret of Their Activity? Int J Mol Sci 2023; 24:ijms24021579. [PMID: 36675102 PMCID: PMC9863470 DOI: 10.3390/ijms24021579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Secreted phospholipases of type A2 (sPLA2s) are proteins of 14-16 kDa present in mammals in different forms and at different body sites. They are involved in lipid transformation processes, and consequently in various immune, inflammatory, and metabolic processes. sPLA2s are also major components of snake venoms, endowed with various toxic and pharmacological properties. The activity of sPLA2s is not limited to the enzymatic one but, through interaction with different types of molecules, they exert other activities that are still little known and explored, both outside and inside the cells, as they can be endocytosed. The aim of this review is to analyze three features of sPLA2s, yet under-explored, knowledge of which could be crucial to understanding the activity of these proteins. The first feature is their disulphide bridge pattern, which has always been considered immutable and necessary for their stability, but which might instead be modulable. The second characteristic is their ability to undergo various post-translational modifications that would control their interaction with other molecules. The third feature is their ability to participate in active molecular condensates both on the surface and within the cell. Finally, the implications of these features in the design of anti-inflammatory drugs are discussed.
Collapse
|
6
|
Kudo K, Miki Y, Carreras J, Nakayama S, Nakamoto Y, Ito M, Nagashima E, Yamamoto K, Higuchi H, Morita SY, Inoue A, Aoki J, Ando K, Nakamura N, Murakami M, Kotani A. Secreted phospholipase A 2 modifies extracellular vesicles and accelerates B cell lymphoma. Cell Metab 2022; 34:615-633.e8. [PMID: 35294862 DOI: 10.1016/j.cmet.2022.02.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 11/15/2021] [Accepted: 02/22/2022] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles (EVs) including exosomes act as intercellular communicators by transferring protein and microRNA cargoes, yet the role of EV lipids remains unclear. Here, we show that the pro-tumorigenic action of lymphoma-derived EVs is augmented via secreted phospholipase A2 (sPLA2)-driven lipid metabolism. Hydrolysis of EV phospholipids by group X sPLA2, which was induced in macrophages of Epstein-Barr virus (EBV) lymphoma, increased the production of fatty acids, lysophospholipids, and their metabolites. sPLA2-treated EVs were smaller and self-aggregated, showed better uptake, and increased cytokine expression and lipid mediator signaling in tumor-associated macrophages. Pharmacological inhibition of endogenous sPLA2 suppressed lymphoma growth in EBV-infected humanized mice, while treatment with sPLA2-modified EVs reversed this phenotype. Furthermore, sPLA2 expression in human large B cell lymphomas inversely correlated with patient survival. Overall, the sPLA2-mediated EV modification promotes tumor development, highlighting a non-canonical mechanistic action of EVs as an extracellular hydrolytic platform of sPLA2.
Collapse
Affiliation(s)
- Kai Kudo
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan; Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental Metabolic Health Sciences Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Joaquim Carreras
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Shunya Nakayama
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan; Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan; Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Yasushi Nakamoto
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan; Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Masatoshi Ito
- Support Center for Medical Research and Education, Tokai University School of Medicine, Isehara, Japan
| | - Etsuko Nagashima
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan; Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Kei Yamamoto
- Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Higuchi
- Center for Cancer Immunology, Cutaneous Biology Research Center, Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shin-Ya Morita
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Japan
| | - Asuka Inoue
- Department of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Kiyoshi Ando
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental Metabolic Health Sciences Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Ai Kotani
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan; Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan.
| |
Collapse
|
7
|
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
|
8
|
Nolin JD, Murphy RC, Gelb MH, Altemeier WA, Henderson WR, Hallstrand TS. Function of secreted phospholipase A 2 group-X in asthma and allergic disease. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:827-837. [PMID: 30529275 DOI: 10.1016/j.bbalip.2018.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
Elevated secreted phospholipase A2 (sPLA2) activity in the airways has been implicated in the pathogenesis of asthma and allergic disease for some time. The identity and function of these enzymes in asthma is becoming clear from work in our lab and others. We focused on sPLA2 group X (sPLA2-X) after identifying increased levels of this enzyme in asthma, and that it is responsible for a large portion of sPLA2 activity in the airways and that the levels are strongly associated with features of airway hyperresponsiveness (AHR). In this review, we discuss studies that implicated sPLA2-X in human asthma, and murine models that demonstrate a critical role of this enzyme as a regulator of type-2 inflammation, AHR and production of eicosanoids. We discuss the mechanism by which sPLA2-X acts to regulate eicosanoids in leukocytes, as well as effects that are mediated through the generation of lysophospholipids and through receptor-mediated functions. This article is part of a Special Issue entitled Novel functions of phospholipase A2 Guest Editors: Makoto Murakami and Gerard Lambeau.
Collapse
Affiliation(s)
- James D Nolin
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of Washington, Seattle, WA, United States of America
| | - Ryan C Murphy
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of Washington, Seattle, WA, United States of America
| | - Michael H Gelb
- Department of Chemistry, University of Washington, Seattle, WA, United States of America; Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | - William A Altemeier
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of Washington, Seattle, WA, United States of America
| | - William R Henderson
- Division of Allergy and Infectious DIseases, University of Washington, Seattle, WA, United States of America
| | - Teal S Hallstrand
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of Washington, Seattle, WA, United States of America.
| |
Collapse
|
9
|
Murakami M, Miki Y, Sato H, Murase R, Taketomi Y, Yamamoto K. Group IID, IIE, IIF and III secreted phospholipase A 2s. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:803-818. [PMID: 30905347 PMCID: PMC7106514 DOI: 10.1016/j.bbalip.2018.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/31/2018] [Accepted: 08/27/2018] [Indexed: 12/02/2022]
Abstract
Among the 11 members of the secreted phospholipase A2 (sPLA2) family, group IID, IIE, IIF and III sPLA2s (sPLA2-IID, -IIE, -IIF and -III, respectively) are “new” isoforms in the history of sPLA2 research. Relative to the better characterized sPLA2s (sPLA2-IB, -IIA, -V and -X), the enzymatic properties, distributions, and functions of these “new” sPLA2s have remained obscure until recently. Our current studies using knockout and transgenic mice for a nearly full set of sPLA2s, in combination with comprehensive lipidomics, have revealed unique and distinct roles of these “new” sPLA2s in specific biological events. Thus, sPLA2-IID is involved in immune suppression, sPLA2-IIE in metabolic regulation and hair follicle homeostasis, sPLA2-IIF in epidermal hyperplasia, and sPLA2-III in male reproduction, anaphylaxis, colonic diseases, and possibly atherosclerosis. In this article, we overview current understanding of the properties and functions of these sPLA2s and their underlying lipid pathways in vivo.
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, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, 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, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Remi Murase
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, 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, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kei Yamamoto
- PRIME, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan; Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan.
| |
Collapse
|
10
|
Yap WH, Ooi BK, Ahmed N, Lim YM. Maslinic acid modulates secreted phospholipase A2-IIA (sPLA2-IIA)-mediated inflammatory effects in macrophage foam cells formation. J Biosci 2018. [DOI: 10.1007/s12038-018-9745-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
11
|
Efficient heterologous expression, functional characterization and molecular modeling of annular seabream digestive phospholipase A2. Chem Phys Lipids 2018. [DOI: 10.1016/j.chemphyslip.2017.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
12
|
Yap WH, Phang SW, Ahmed N, Lim YM. Differential effects of sPLA 2-GV and GX on cellular proliferation and lipid accumulation in HT29 colon cancer cells. Mol Cell Biochem 2018; 447:93-101. [PMID: 29374817 DOI: 10.1007/s11010-018-3295-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/23/2018] [Indexed: 11/29/2022]
Abstract
Secretory phospholipase A2 (sPLA2) group of enzymes have been shown to hydrolyze phospholipids, among which sPLA2 Group V (GV) and Group X (GX) exhibit high selectivity towards phosphatidylcholine-rich cellular plasma membranes. The enzymes have recently emerged as key regulators in lipid droplets formation and it is hypothesized that sPLA2-GV and GX enhanced cell proliferation and lipid droplet accumulation in colon cancer cells (HT29). In this study, cell viability and lipid droplet accumulation were assessed by Resazurin assay and Oil-Red-O staining. Interestingly, both sPLA2-GV and GX enzymes reduced intracellular lipid droplet accumulation and did not significantly affect cell proliferation in HT29 cells. Incubation with varespladib, a pan-inhibitor of sPLA2-Group IIA/V/X, further suppressed lipid droplets accumulation in sPLA2-GV but have no effects in sPLA2-GX-treated cells. Further studies using catalytically inactive sPLA2 enzymes showed that the enzymes intrinsic catalytic activity is required for the net reduction of lipid accumulation. Meanwhile, inhibition of intracellular phospholipases (iPLA2-γ and cPLA2-α) unexpectedly enhanced lipid droplet accumulation in both sPLA2-GV and GX-treated cells. The findings suggested an interconnected relationship between extracellular and intracellular phospholipases in lipid cycling. Previous studies indicated that sPLA2 enzymes are linked to cancer development due to their ability to induce release of arachidonic acid and eicosanoids as well as the stimulation of lipid droplet formation. This study showed that the two enzymes work in a distinct manner and they neither confer proliferative advantage nor enhanced the net lipid droplet accumulation in HT29 cells.
Collapse
Affiliation(s)
- Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Selangor, Malaysia.
| | - Su Wen Phang
- School of Biosciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Nafees Ahmed
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Yang Mooi Lim
- Department of Pre-clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Selangor, Malaysia
| |
Collapse
|
13
|
Nedvedova I, Kolar D, Neckar J, Kalous M, Pravenec M, Šilhavý J, Korenkova V, Kolar F, Zurmanova JM. Cardioprotective Regimen of Adaptation to Chronic Hypoxia Diversely Alters Myocardial Gene Expression in SHR and SHR-mt BN Conplastic Rat Strains. Front Endocrinol (Lausanne) 2018; 9:809. [PMID: 30723458 PMCID: PMC6350269 DOI: 10.3389/fendo.2018.00809] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/24/2018] [Indexed: 11/17/2022] Open
Abstract
Adaptation to continuous normobaric hypoxia (CNH) protects the heart against acute ischemia/reperfusion injury. Recently, we have demonstrated the infarct size-limiting effect of CNH also in hearts of spontaneously hypertensive rats (SHR) and in conplastic SHR-mtBN strain characterized by the selective replacement of the mitochondrial genome of SHR with that of more ischemia-resistant Brown Norway rats. Importantly, cardioprotective effect of CNH was more pronounced in SHR-mtBN than in SHR. Thus, here we aimed to identify candidate genes which may contribute to this difference between the strains. Rats were adapted to CNH (FiO2 0.1) for 3 weeks or kept at room air as normoxic controls. Screening of 45 transcripts was performed in left ventricles using Biomark Chip. Significant differences between the groups were analyzed by univariate analysis (ANOVA) and the genes contributing to the differences between the strains unmasked by CNH were identified by multivariate analyses (PCA, SOM). ANOVA with Bonferroni correction revealed that transcripts differently affected by CNH in SHR and SHR-mtBN belong predominantly to lipid metabolism and antioxidant defense. PCA divided four experimental groups into two main clusters corresponding to chronically hypoxic and normoxic groups, and differences between the strains were more pronounced after CNH. Subsequently, the following 14 candidate transcripts were selected by PCA, and confirmed by SOM analyses, that can contribute to the strain differences in cardioprotective phenotype afforded by CNH: Alkaline ceramidase 2 (Acer2), Fatty acid translocase (Cd36), Aconitase 1 (Aco1), Peroxisome proliferator activated receptor gamma (Pparg), Hemoxygenase 2 (Hmox2), Phospholipase A2 group IIA (Ppla2g2a), Dynamin-related protein (Drp), Protein kinase C epsilon (Pkce), Hexokinase 2 (Hk2), Sphingomyelin synthase 2 (Sgms2), Caspase 3 (Casp3), Mitofussin 1 (Mfn1), Phospholipase A2 group V (Pla2g5), and Catalase (Cat). Our data suggest that the stronger cardioprotective phenotype of conplastic SHR-mtBN strain afforded by CNH is associated with either preventing the drop or increasing the expression of transcripts related to energy metabolism, antioxidant response and mitochondrial dynamics.
Collapse
Affiliation(s)
- Iveta Nedvedova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - David Kolar
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Jan Neckar
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Martin Kalous
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Jan Šilhavý
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Vlasta Korenkova
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czechia
| | - Frantisek Kolar
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Jitka M. Zurmanova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
- *Correspondence: Jitka M. Zurmanova
| |
Collapse
|
14
|
Hydrolysis of Phosphatidylcholine-Isoprostanes (PtdCho-IP) by Peripheral Human Group IIA, V and X Secretory Phospholipases A2 (sPLA2). Lipids 2017; 52:477-488. [DOI: 10.1007/s11745-017-4264-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/08/2017] [Indexed: 10/19/2022]
|
15
|
Ghomashchi F, Brglez V, Payré C, Jeammet L, Bezzine S, Gelb MH, Lambeau G. Preparation of the Full Set of Recombinant Mouse- and Human-Secreted Phospholipases A 2. Methods Enzymol 2016; 583:35-69. [PMID: 28063498 DOI: 10.1016/bs.mie.2016.10.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A family of 14-20kDa, disulfide-rich, calcium-dependent secreted phospholipases A2 (sPLA2s) that release fatty acids from the sn-2 position of glycerophospholipids can be found in mammals. They have a diverse array of tissue distribution and biological functions. In this chapter we provide detailed protocols for production of nearly all of the mouse and human sPLA2s mainly by expression in bacteria and in vitro refolding or by expression in insect cells. High-resolution mass spectrometry and enzymatic assays were, respectively, used to show that all disulfides are formed and that the enzymes are active, strongly suggesting that each sPLA2 was prepared in the structurally native form. The availability of these proteins has allowed kinetic studies to be carried out, to prepare highly selective antisera, to screen for selective inhibitors, to study receptor binding, and to study the action of each enzyme on mammalian cell membranes and their in vivo biological roles.
Collapse
Affiliation(s)
- F Ghomashchi
- University of Washington, Seattle, WA, United States
| | - V Brglez
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia Antipolis and Centre National de la Recherche Scientifique (CNRS), Valbonne, France
| | - C Payré
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia Antipolis and Centre National de la Recherche Scientifique (CNRS), Valbonne, France
| | - L Jeammet
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia Antipolis and Centre National de la Recherche Scientifique (CNRS), Valbonne, France
| | - S Bezzine
- University of Sfax, Laboratoire de Biochimie et de Génie Enzymatique des Lipases, Sfax, Tunisia
| | - M H Gelb
- University of Washington, Seattle, WA, United States.
| | - G Lambeau
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia Antipolis and Centre National de la Recherche Scientifique (CNRS), Valbonne, France.
| |
Collapse
|
16
|
Murakami M, Yamamoto K, Miki Y, Murase R, Sato H, Taketomi Y. The Roles of the Secreted Phospholipase A 2 Gene Family in Immunology. Adv Immunol 2016; 132:91-134. [PMID: 27769509 PMCID: PMC7112020 DOI: 10.1016/bs.ai.2016.05.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Within the phospholipase A2 (PLA2) family that hydrolyzes phospholipids to yield fatty acids and lysophospholipids, secreted PLA2 (sPLA2) enzymes comprise the largest group containing 11 isoforms in mammals. Individual sPLA2s exhibit unique tissue or cellular distributions and enzymatic properties, suggesting their distinct biological roles. Although PLA2 enzymes, particularly cytosolic PLA2 (cPLA2α), have long been implicated in inflammation by driving arachidonic acid metabolism, the precise biological roles of sPLA2s have remained a mystery over the last few decades. Recent studies employing mice gene-manipulated for individual sPLA2s, in combination with mass spectrometric lipidomics to identify their target substrates and products in vivo, have revealed their roles in diverse biological events, including immunity and associated disorders, through lipid mediator-dependent or -independent processes in given microenvironments. In this review, we summarize our current knowledge of the roles of sPLA2s in various immune responses and associated diseases.
Collapse
Affiliation(s)
- M Murakami
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| | - K Yamamoto
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Y Miki
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - R Murase
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - H Sato
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Y Taketomi
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
17
|
Savas S, Kabaroglu C, Alpman A, Sarac F, Yalcin MA, Parıldar Z, Ozkinay F, Kumral E, Akcicek F. No relationship between lipoprotein-associated phospholipase A2, proinflammatory cytokines, and neopterin in Alzheimer's disease. Exp Gerontol 2016; 77:1-6. [PMID: 26828804 DOI: 10.1016/j.exger.2016.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Lipoprotein-associated phospholipase A2 (Lp-PLA2) is a reported risk factor for dementia. However, the relationship between Alzheimer's disease (AD) and Lp-PLA2 is still debatable and, to the best of our knowledge, no study has evaluated the associations between levels of Lp-PLA2, proinflammatory cytokines, and neopterin in AD. METHODS In total, 59 patients with AD and 38 non-demented individuals were included in the case-control study. Fasting serum concentrations of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), neopterin, and Lp-PLA2 were determined using ELISA. The associations between AD and each of the variables were analyzed by logistic regression. RESULTS The median Lp-PLA2 levels in AD and controls were similar (P=0.29, not significant). Median serum neopterin and IL-6 levels were significantly higher in patients with AD than in controls (P=0.0001 and P=0.03, respectively). In regression analyses, median neopterin levels, a lower level of education, and female gender were significantly associated with AD when compared with controls (OR, 31.44, 95% CI 3.59-275.28, P=0.002; OR, 4.35, 95% CI 1.13-16.61, P=0.032; OR, 7.25, 95% CI 1.88-28.00, P=0.004, respectively). CONCLUSION In contrast to previous evidence suggesting its role in dementia and AD, Lp-PLA2 enzyme levels were higher in the controls, and no relationship between Lp-PLA2 and either proinflammatory cytokines or neopterin was identified in AD. Elevated neopterin levels may be considered inflammatory markers of AD.
Collapse
Affiliation(s)
- S Savas
- Division of Geriatric Medicine, Department of Internal Medicine, School of Medicine, Ege University, Izmir, Turkey.
| | - C Kabaroglu
- Department of Clinical Biochemistry, School of Medicine, Ege University, Izmir, Turkey
| | - A Alpman
- Department of Medical Genetics, School of Medicine, Ege University, Izmir, Turkey
| | - F Sarac
- Division of Geriatric Medicine, Department of Internal Medicine, School of Medicine, Ege University, Izmir, Turkey
| | - M A Yalcin
- Division of Geriatric Medicine, Department of Internal Medicine, School of Medicine, Ege University, Izmir, Turkey
| | - Z Parıldar
- Department of Clinical Biochemistry, School of Medicine, Ege University, Izmir, Turkey
| | - F Ozkinay
- Department of Medical Genetics, School of Medicine, Ege University, Izmir, Turkey
| | - E Kumral
- Department of Neurology, School of Medicine, Ege University, Izmir, Turkey
| | - F Akcicek
- Division of Geriatric Medicine, Department of Internal Medicine, School of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
18
|
Endogenous secreted phospholipase A2 group X regulates cysteinyl leukotrienes synthesis by human eosinophils. J Allergy Clin Immunol 2015; 137:268-277.e8. [PMID: 26139511 DOI: 10.1016/j.jaci.2015.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 04/23/2015] [Accepted: 05/04/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Phospholipase A2s mediate the rate-limiting step in the formation of eicosanoids such as cysteinyl leukotrienes (CysLTs). Group IVA cytosolic PLA2α (cPLA2α) is thought to be the dominant PLA2 in eosinophils; however, eosinophils also have secreted PLA2 (sPLA2) activity that has not been fully defined. OBJECTIVES To examine the expression of sPLA2 group X (sPLA2-X) in eosinophils, the participation of sPLA2-X in the formation of CysLTs, and the mechanism by which sPLA2-X initiates the synthesis of CysLTs in eosinophils. METHODS Peripheral blood eosinophils were obtained from volunteers with asthma and/or allergy. A rabbit polyclonal anti-sPLA2-X antibody identified sPLA2-X by Western blot. We used confocal microscopy to colocalize the sPLA2-X to intracellular structures. An inhibitor of sPLA2-X (ROC-0929) that does not inhibit other mammalian sPLA2s, as well as inhibitors of the mitogen-activated kinase cascade (MAPK) and cPLA2α, was used to examine the mechanism of N-formyl-methionyl-leucyl-phenylalanine (fMLP)-mediated formation of CysLT. RESULTS Eosinophils express the mammalian sPLA2-X gene (PLA2G10). The sPLA2-X protein is located in the endoplasmic reticulum, golgi, and granules of eosinophils and moves to the granules and lipid bodies during fMLP-mediated activation. Selective sPLA2-X inhibition attenuated the fMLP-mediated release of arachidonic acid and CysLT formation by eosinophils. Inhibitors of p38, extracellular-signal-regulated kinases 1/2 (p44/42 MAPK), c-Jun N-terminal kinase, and cPLA2α also attenuated the fMLP-mediated formation of CysLT. The sPLA2-X inhibitor reduced the phosphorylation of p38 and extracellular-signal-regulated kinases 1/2 (p44/42 MAPK) as well as cPLA2α during cellular activation, indicating that sPLA2-X is involved in activating the MAPK cascade leading to the formation of CysLT via cPLA2α. We further demonstrate that sPLA2-X is activated before secretion from the cell during activation. Short-term priming with IL-13 and TNF/IL-1β increased the expression of PLA2G10 by eosinophils. CONCLUSIONS These results demonstrate that sPLA2-X plays a significant role in the formation of CysLTs by human eosinophils. The predominant role of the enzyme is the regulation of MAPK activation that leads to the phosphorylation of cPLA2α. The sPLA2-X protein is regulated by proteolytic cleavage, suggesting that an inflammatory environment may promote the formation of CysLTs through this mechanism. These results have important implications for the treatment of eosinophilic disorders such as asthma.
Collapse
|
19
|
Rousseau M, Naika GS, Perron J, Jacques F, Gelb MH, Boilard E. Study of the role of cytosolic phospholipase A2 alpha in eicosanoid generation and thymocyte maturation in the thymus. PLoS One 2015; 10:e0126204. [PMID: 25969996 PMCID: PMC4430275 DOI: 10.1371/journal.pone.0126204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 03/30/2015] [Indexed: 11/29/2022] Open
Abstract
The thymus is a primary lymphoid organ, home of maturation and selection of thymocytes for generation of functional T-cells. Multiple factors are involved throughout the different stages of the maturation process to tightly regulate T-cell production. The metabolism of arachidonic acid by cyclooxygenases, lipoxygenases and specific isomerases generates eicosanoids, lipid mediators capable of triggering cellular responses. In this study, we determined the profile of expression of the eicosanoids present in the mouse thymus at different stages of thymocyte development. As the group IVA cytosolic phospholipase A2 (cPLA2α) catalyzes the hydrolysis of phospholipids, thereby generating arachidonic acid, we further verified its contribution by including cPLA2α deficient mice to our investigations. We found that a vast array of eicosanoids is expressed in the thymus, which expression is substantially modulated through thymocyte development. The cPLA2α was dispensable in the generation of most eicosanoids in the thymus and consistently, the ablation of the cPLA2α gene in mouse thymus and the culture of thymuses from human newborns in presence of the cPLA2α inhibitor pyrrophenone did not impact thymocyte maturation. This study provides information on the eicosanoid repertoire present during thymocyte development and suggests that thymocyte maturation can occur independently of cPLA2α.
Collapse
Affiliation(s)
- Matthieu Rousseau
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Gajendra S. Naika
- Department of Chemistry, University of Washington, Seattle, WA, the United States of America
| | - Jean Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Frederic Jacques
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, WA, the United States of America
| | - Eric Boilard
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
- * E-mail:
| |
Collapse
|
20
|
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
|
21
|
Layne JD, Shridas P, Webb NR. Ectopically expressed pro-group X secretory phospholipase A2 is proteolytically activated in mouse adrenal cells by furin-like proprotein convertases: implications for the regulation of adrenal steroidogenesis. J Biol Chem 2015; 290:7851-60. [PMID: 25623068 DOI: 10.1074/jbc.m114.634667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Group X secretory phospholipase A2 (GX sPLA2) hydrolyzes mammalian cell membranes, liberating free fatty acids and lysophospholipids. GX sPLA2 is produced as a pro-enzyme (pro-GX sPLA2) that contains an N-terminal 11-amino acid propeptide ending in a dibasic motif, suggesting cleavage by a furin-like proprotein convertase (PC). Although propeptide cleavage is clearly required for enzymatic activity, the protease(s) responsible for pro-GX sPLA2 activation have not been identified. We previously reported that GX sPLA2 negatively regulates adrenal glucocorticoid production, likely by suppressing liver X receptor-mediated activation of steroidogenic acute regulatory protein expression. In this study, using a FLAG epitope-tagged pro-GX sPLA2 expression construct (FLAG-pro-GX sPLA2), we determined that adrenocorticotropic hormone (ACTH) enhanced FLAG-pro-GX sPLA2 processing and phospholipase activity secreted by Y1 adrenal cells. ACTH increased the expression of furin and PCSK6, but not other members of the PC family, in Y1 cells. Overexpression of furin and PCSK6 in HEK 293 cells significantly enhanced FLAG-pro-GX sPLA2 processing, whereas siRNA-mediated knockdown of both PCs almost completely abolished FLAG-pro-GX sPLA2 processing in Y1 cells. Expression of either furin or PCSK6 enhanced the ability of GX sPLA2 to suppress liver X receptor reporter activity. The PC inhibitor decanoyl-Arg-Val-Lys-Arg-chloromethyl ketone significantly suppressed FLAG-pro-GX sPLA2 processing and sPLA2 activity in Y1 cells, and it significantly attenuated GX sPLA2-dependent inhibition of steroidogenic acute regulatory protein expression and progesterone production. These findings provide strong evidence that pro-GX sPLA2 is a substrate for furin and PCSK6 proteolytic processing and define a novel mechanism for regulating corticosteroid production in adrenal cells.
Collapse
Affiliation(s)
- Joseph D Layne
- From the Department of Pharmacology and Nutritional Sciences, Division of Nutritional Sciences, the Saha Cardiovascular Research Center, and
| | - Preetha Shridas
- the Department of Internal Medicine, University of Kentucky Medical Center, Lexington, Kentucky 40536
| | - Nancy R Webb
- the Department of Internal Medicine, University of Kentucky Medical Center, Lexington, Kentucky 40536
| |
Collapse
|
22
|
Rousseau M, Belleannee C, Duchez AC, Cloutier N, Levesque T, Jacques F, Perron J, Nigrovic PA, Dieude M, Hebert MJ, Gelb MH, Boilard E. Detection and quantification of microparticles from different cellular lineages using flow cytometry. Evaluation of the impact of secreted phospholipase A2 on microparticle assessment. PLoS One 2015; 10:e0116812. [PMID: 25587983 PMCID: PMC4294685 DOI: 10.1371/journal.pone.0116812] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 12/15/2014] [Indexed: 11/20/2022] Open
Abstract
Microparticles, also called microvesicles, are submicron extracellular vesicles produced by plasma membrane budding and shedding recognized as key actors in numerous physio(patho)logical processes. Since they can be released by virtually any cell lineages and are retrieved in biological fluids, microparticles appear as potent biomarkers. However, the small dimensions of microparticles and soluble factors present in body fluids can considerably impede their quantification. Here, flow cytometry with improved methodology for microparticle resolution was used to detect microparticles of human and mouse species generated from platelets, red blood cells, endothelial cells, apoptotic thymocytes and cells from the male reproductive tract. A family of soluble proteins, the secreted phospholipases A2 (sPLA2), comprises enzymes concomitantly expressed with microparticles in biological fluids and that catalyze the hydrolysis of membrane phospholipids. As sPLA2 can hydrolyze phosphatidylserine, a phospholipid frequently used to assess microparticles, and might even clear microparticles, we further considered the impact of relevant sPLA2 enzymes, sPLA2 group IIA, V and X, on microparticle quantification. We observed that if enriched in fluids, certain sPLA2 enzymes impair the quantification of microparticles depending on the species studied, the source of microparticles and the means of detection employed (surface phosphatidylserine or protein antigen detection). This study provides analytical considerations for appropriate interpretation of microparticle cytofluorometric measurements in biological samples containing sPLA2 enzymes.
Collapse
Affiliation(s)
- Matthieu Rousseau
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Clemence Belleannee
- Centre de Recherche du CHUQ and Département d’Obstétrique-Gynécologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Anne-Claire Duchez
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Nathalie Cloutier
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | - Tania Levesque
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
| | | | - Jean Perron
- Centre Hospitalier Universitaire de Québec, Québec, Canada
| | - Peter A. Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Melanie Dieude
- Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Marie-Josee Hebert
- Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, WA, United States of America
| | - Eric Boilard
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l’Université Laval, Québec, QC, Canada
- * E-mail:
| |
Collapse
|
23
|
Shridas P, Zahoor L, Forrest KJ, Layne JD, Webb NR. Group X secretory phospholipase A2 regulates insulin secretion through a cyclooxygenase-2-dependent mechanism. J Biol Chem 2014; 289:27410-7. [PMID: 25122761 DOI: 10.1074/jbc.m114.591735] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Group X secretory phospholipase A2 (GX sPLA2) potently hydrolyzes membrane phospholipids to release arachidonic acid (AA). While AA is an activator of glucose-stimulated insulin secretion (GSIS), its metabolite prostaglandin E2 (PGE2) is a known inhibitor. In this study, we determined that GX sPLA2 is expressed in insulin-producing cells of mouse pancreatic islets and investigated its role in beta cell function. GSIS was measured in vivo in wild-type (WT) and GX sPLA2-deficient (GX KO) mice and ex vivo using pancreatic islets isolated from WT and GX KO mice. GSIS was also assessed in vitro using mouse MIN6 pancreatic beta cells with or without GX sPLA2 overexpression or exogenous addition. GSIS was significantly higher in islets isolated from GX KO mice compared with islets from WT mice. Conversely, GSIS was lower in MIN6 cells overexpressing GX sPLA2 (MIN6-GX) compared with control (MIN6-C) cells. PGE2 production was significantly higher in MIN6-GX cells compared with MIN6-C cells and this was associated with significantly reduced cellular cAMP. The effect of GX sPLA2 on GSIS was abolished when cells were treated with NS398 (a COX-2 inhibitor) or L-798,106 (a PGE2-EP3 receptor antagonist). Consistent with enhanced beta cell function, GX KO mice showed significantly increased plasma insulin levels following glucose challenge and were protected from age-related reductions in GSIS and glucose tolerance compared with WT mice. We conclude that GX sPLA2 plays a previously unrecognized role in negatively regulating pancreatic insulin secretion by augmenting COX-2-dependent PGE2 production.
Collapse
Affiliation(s)
- Preetha Shridas
- From Saha Cardiovascular Research Center and Departments of Internal Medicine and
| | - Lubna Zahoor
- From Saha Cardiovascular Research Center and Departments of Internal Medicine and
| | - Kathy J Forrest
- From Saha Cardiovascular Research Center and Departments of Internal Medicine and
| | - Joseph D Layne
- From Saha Cardiovascular Research Center and Pharmacology and Nutritional Sciences, Division of Nutritional Sciences, University of Kentucky Medical Center, Lexington Kentucky 40536
| | - Nancy R Webb
- From Saha Cardiovascular Research Center and Pharmacology and Nutritional Sciences, Division of Nutritional Sciences, University of Kentucky Medical Center, Lexington Kentucky 40536
| |
Collapse
|
24
|
The (G>A) rs11573191 polymorphism of PLA2G5 gene is associated with premature coronary artery disease in the Mexican Mestizo population: the genetics of atherosclerotic disease Mexican study. BIOMED RESEARCH INTERNATIONAL 2014; 2014:931361. [PMID: 24959594 PMCID: PMC4052156 DOI: 10.1155/2014/931361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/02/2014] [Accepted: 05/04/2014] [Indexed: 11/20/2022]
Abstract
Coronary artery disease (CAD) is a multifactorial disorder that results from an excessive inflammatory response. Secretory phospholipase A2-V (sPLA2-V) encoded by PLA2G5 gene promotes diverse proinflammatory processes. The aim of the present study was to analyze if PLA2G5 gene polymorphisms are associated with premature CAD. Three PLA2G5 polymorphisms (rs11573187, rs2148911, and rs11573191) were analyzed in 707 patients with premature CAD and 749 healthy controls. Haplotypes were constructed after linkage disequilibrium analysis. Under dominant, recessive, and additive models, the rs11573191 polymorphism was associated with increased risk of premature CAD (OR = 1.51, Pdom = 3.5 × 10−3; OR = 2.95, Prec = 0.023; OR = 1.51, Padd = 1.2 × 10−3). According to the informatics software, this polymorphism had a functional effect modifying the affinity of the sequence by the MZF1 transcription factor. PLA2G5 polymorphisms were in linkage disequilibrium and the CGA haplotype was associated with increased risk of premature CAD (OR = 1.49, P = 0.0023) and with hypertension in these patients (OR = 1.75, P = 0.0072). Our results demonstrate the association of the PLA2G5 rs11573191 polymorphism with premature CAD. In our study, it was possible to distinguish one haplotype associated with increased risk of premature CAD and hypertension.
Collapse
|
25
|
Nyegaard S, Novakovic VA, Rasmussen JT, Gilbert GE. Lactadherin inhibits secretory phospholipase A2 activity on pre-apoptotic leukemia cells. PLoS One 2013; 8:e77143. [PMID: 24194865 PMCID: PMC3806724 DOI: 10.1371/journal.pone.0077143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/21/2013] [Indexed: 01/09/2023] Open
Abstract
Secretory phospholipase A2 (sPLA2) is a critical component of insect and snake venoms and is secreted by mammalian leukocytes during inflammation. Elevated secretory PLA2 concentrations are associated with autoimmune diseases and septic shock. Many sPLA2’s do not bind to plasma membranes of quiescent cells but bind and digest phospholipids on the membranes of stimulated or apoptotic cells. The capacity of these phospholipases to digest membranes of stimulated or apoptotic cells correlates to the exposure of phosphatidylserine. In the present study, the ability of the phosphatidyl-L-serine-binding protein, lactadherin to inhibit phospholipase enzyme activity has been assessed. Inhibition of human secretory phospholipase A2-V on phospholipid vesicles exceeded 90%, whereas inhibition of Naja mossambica sPLA2 plateaued at 50–60%. Lactadherin inhibited 45% of activity of Naja mossambica sPLA2 and >70% of human secretory phospholipase A2-V on the membranes of human NB4 leukemia cells treated with calcium ionophore A23187. The data indicate that lactadherin may decrease inflammation by inhibiting sPLA2.
Collapse
Affiliation(s)
- Steffen Nyegaard
- Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
- Departments of Medicine, Veterans Administration Boston Healthcare System, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Valerie A. Novakovic
- Departments of Medicine, Veterans Administration Boston Healthcare System, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jan T. Rasmussen
- Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
- * E-mail:
| | - Gary E. Gilbert
- Departments of Medicine, Veterans Administration Boston Healthcare System, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
26
|
The role of secretory phospholipase A₂ in the central nervous system and neurological diseases. Mol Neurobiol 2013; 49:863-76. [PMID: 24113843 DOI: 10.1007/s12035-013-8565-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/25/2013] [Indexed: 12/31/2022]
Abstract
Secretory phospholipase A2 (sPLA2s) are small secreted proteins (14-18 kDa) and require submillimolar levels of Ca(2+) for liberating arachidonic acid from cell membrane lipids. In addition to the enzymatic function, sPLA2 can exert various biological responses by binding to specific receptors. Physiologically, sPLA2s play important roles on the neurotransmission in the central nervous system and the neuritogenesis in the peripheral nervous system. Pathologically, sPLA2s are involved in the neurodegenerative diseases (e.g., Alzheimer's disease) and cerebrovascular diseases (e.g., stoke). The common pathology (e.g., neuronal apoptosis) of Alzheimer's disease and stroke coexists in the mixed dementia, suggesting common pathogenic mechanisms of the two neurological diseases. Among mammalian sPLA2s, sPLA2-IB and sPLA2-IIA induce neuronal apoptosis in rat cortical neurons. The excess influx of calcium into neurons via L-type voltage-dependent Ca(2+) channels mediates the two sPLA2-induced apoptosis. The elevated concentration of intracellular calcium activates PKC, MAPK and cytosolic PLA2. Moreover, it is linked with the production of reactive oxygen species and apoptosis through activation of the superoxide producing enzyme NADPH oxidase. NADPH oxidase is involved in the neurotoxicity of amyloid β peptide, which impairs synaptic plasticity long before its deposition in the form of amyloid plaques of Alzheimer's disease. In turn, reactive oxygen species from NADPH oxidase can stimulate ERK1/2 phosphorylation and activation of cPLA2 and result in a release of arachidonic acid. sPLA2 is up-regulated in both Alzheimer's disease and cerebrovascular disease, suggesting the involvement of sPLA2 in the common pathogenic mechanisms of the two diseases. Thus, our review presents evidences for pathophysiological roles of sPLA2 in the central nervous system and neurological diseases.
Collapse
|
27
|
Pucer A, Brglez V, Payré C, Pungerčar J, Lambeau G, Petan T. Group X secreted phospholipase A(2) induces lipid droplet formation and prolongs breast cancer cell survival. Mol Cancer 2013; 12:111. [PMID: 24070020 PMCID: PMC3852912 DOI: 10.1186/1476-4598-12-111] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 09/24/2013] [Indexed: 01/08/2023] Open
Abstract
Background Alterations in lipid metabolism are inherent to the metabolic transformations that support tumorigenesis. The relationship between the synthesis, storage and use of lipids and their importance in cancer is poorly understood. The human group X secreted phospholipase A2 (hGX sPLA2) releases fatty acids (FAs) from cell membranes and lipoproteins, but its involvement in the regulation of cellular FA metabolism and cancer is not known. Results Here we demonstrate that hGX sPLA2 induces lipid droplet (LD) formation in invasive breast cancer cells, stimulates their proliferation and prevents their death on serum deprivation. The effects of hGX sPLA2 are shown to be dependent on its enzymatic activity, are mimicked by oleic acid and include activation of protein kinase B/Akt, a cell survival signaling kinase. The hGX sPLA2-stimulated LD biogenesis is accompanied by AMP-activated protein kinase (AMPK) activation, up-regulation of FA oxidation enzymes and the LD-coating protein perilipin 2, and suppression of lipogenic gene expression. Prolonged activation of AMPK inhibited hGX sPLA2-induced LD formation, while etomoxir, an inhibitor of FA oxidation, abrogated both LD formation and cell survival. The hGX sPLA2-induced changes in lipid metabolism provide a minimal immediate proliferative advantage during growth under optimal conditions, but they confer to the breast cancer cells a sustained ability to resist apoptosis during nutrient and growth factor limitation. Conclusion Our results identify hGX sPLA2 as a novel modulator of lipid metabolism that promotes breast cancer cell growth and survival by stimulating LD formation and FA oxidation.
Collapse
Affiliation(s)
- Anja Pucer
- Department of Molecular and Biomedical Sciences, JoŽef Stefan Institute, Ljubljana, Slovenia.
| | | | | | | | | | | |
Collapse
|
28
|
Hallstrand TS, Lai Y, Altemeier WA, Appel CL, Johnson B, Frevert CW, Hudkins KL, Bollinger JG, Woodruff PG, Hyde DM, Henderson WR, Gelb MH. Regulation and function of epithelial secreted phospholipase A2 group X in asthma. Am J Respir Crit Care Med 2013; 188:42-50. [PMID: 23614662 PMCID: PMC3735246 DOI: 10.1164/rccm.201301-0084oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/08/2013] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Indirect airway hyperresponsiveness (AHR) is a fundamental feature of asthma that is manifest as exercise-induced bronchoconstriction (EIB). Secreted phospholipase A2 group X (sPLA2-X) plays a key role in regulating eicosanoid formation and the development of inflammation and AHR in murine models. OBJECTIVES We sought to examine sPLA2-X in the airway epithelium and airway wall of patients with asthma, the relationship to AHR in humans, and the regulation and function of sPLA2-X within the epithelium. METHODS We precisely phenotyped 34 patients with asthma (19 with and 15 without EIB) and 10 normal control subjects to examine in vivo differences in epithelial gene expression, quantitative morphometry of endobronchial biopsies, and levels of secreted protein. The regulation of sPLA2-X gene (PLA2G10) expression was examined in primary airway epithelial cell cultures. The function of epithelial sPLA2-X in eicosanoid formation was examined using PLA2 inhibitors and murine tracheal epithelial cells with Pla2g10 deletion. MEASUREMENTS AND MAIN RESULTS We found that sPLA2-X protein is increased in the airways of patients with asthma and that epithelial-derived sPLA2-X may be increased in association with indirect AHR. The expression of sPLA2-X increases during in vitro epithelial differentiation; is regulated by inflammatory signals including tumor necrosis factor, IL-13, and IL-17; and is both secreted from the epithelium and directly participates in the release of arachidonic acid by epithelial cells. CONCLUSIONS These data reveal a relationship between epithelial-derived sPLA2-X and indirect AHR in asthma and that sPLA2-X serves as an epithelial regulator of inflammatory eicosanoid formation. Therapies targeting epithelial sPLA2-X may be useful in asthma.
Collapse
|
29
|
Ait-Oufella H, Herbin O, Lahoute C, Coatrieux C, Loyer X, Joffre J, Laurans L, Ramkhelawon B, Blanc-Brude O, Karabina S, Girard CA, Payré C, Yamamoto K, Binder CJ, Murakami M, Tedgui A, Lambeau G, Mallat Z. Group X Secreted Phospholipase A2 Limits the Development of Atherosclerosis in LDL Receptor–Null Mice. Arterioscler Thromb Vasc Biol 2013; 33:466-73. [DOI: 10.1161/atvbaha.112.300309] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Several secreted phospholipases A2 (sPLA2s), including group IIA, III, V, and X, have been linked to the development of atherosclerosis, which led to the clinical testing of A-002 (varespladib), a broad sPLA2 inhibitor for the treatment of coronary artery disease. Group X sPLA2 (PLA2G10) has the most potent hydrolyzing activity toward phosphatidylcholine and is believed to play a proatherogenic role.
Methods and Results—
Here, we show that
Ldlr
–/–
mice reconstituted with bone marrow from mouse group X–deficient mice (
Pla2g10
–/–
) unexpectedly display a doubling of plaque size compared with
Pla2g10
+/+
chimeric mice. Macrophages of
Pla2g10
–/–
mice are more susceptible to apoptosis in vitro, which is associated with a 4-fold increase of plaque necrotic core in vivo. In addition, chimeric
Pla2g10
–/–
mice show exaggerated T lymphocyte (Th)1 immune response, associated with enhanced T-cell infiltration in atherosclerotic plaques. Interestingly, overexpression of human PLA2G10 in murine bone marrow cells leads to significant reduction of Th1 response and to 50% reduction of lesion size.
Conclusion—
PLA2G10 expression in bone marrow cells controls a proatherogenic Th1 response and limits the development of atherosclerosis. The results may provide an explanation for the recently reported inefficacy of A-002 (varespladib) to treat patients with coronary artery disease. Indeed, A-002 is a nonselective sPLA2 inhibitor that inhibits both proatherogenic (groups IIA and V) and antiatherogenic (group X) sPLA2s. Our results suggest that selective targeting of individual sPLA2 enzymes may be a better strategy to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Olivier Herbin
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Charlotte Lahoute
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christelle Coatrieux
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Xavier Loyer
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Jeremie Joffre
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Ludivine Laurans
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Bhama Ramkhelawon
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Olivier Blanc-Brude
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Sonia Karabina
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christophe A. Girard
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christine Payré
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Kei Yamamoto
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Christoph J. Binder
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Makoto Murakami
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Alain Tedgui
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Gérard Lambeau
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| | - Ziad Mallat
- From the Inserm U970, Paris Cardiovascular Research Center, Université René Descartes, Paris, France (H.A.-O., O.H., C.L., X.L., J.J., L.L., B.R., O.B.-B., A.T., Z.M.); Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Université Pierre et Marie Curie, Paris, France (H.A.-O.); Institute of Molecular and Cellular Pharmacology (IPMC), UMR 7275 CNRS- and Université de Nice-Sophia Antipolis, Valbonne, France (C.C., C.A.G., C.P., G.L.); Inserm UMRS 937, Paris, France (S.K.); Lipid Metabolism
| |
Collapse
|
30
|
Point V, Bénarouche A, Jemel I, Parsiegla G, Lambeau G, Carrière F, Cavalier JF. Effects of the propeptide of group X secreted phospholipase A2 on substrate specificity and interfacial activity on phospholipid monolayers. Biochimie 2013; 95:51-8. [DOI: 10.1016/j.biochi.2012.07.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 07/27/2012] [Indexed: 10/28/2022]
|
31
|
Boyanovsky BB, Bailey W, Dixon L, Shridas P, Webb NR. Group V secretory phospholipase A2 enhances the progression of angiotensin II-induced abdominal aortic aneurysms but confers protection against angiotensin II-induced cardiac fibrosis in apoE-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1088-98. [PMID: 22813854 DOI: 10.1016/j.ajpath.2012.05.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 05/02/2012] [Accepted: 05/17/2012] [Indexed: 01/23/2023]
Abstract
Abdominal aortic aneurysms (AAAs) and heart failure are complex life-threatening diseases whose etiology is not completely understood. In this study, we investigated whether deficiency of group V secretory phospholipase A(2) (GV sPLA(2)) protects from experimental AAA. The impact of GV sPLA(2) deficiency on angiotensin (Ang) II-induced cardiac fibrosis was also investigated. Apolipoprotein E (apoE)(-/-) mice and apoE(-/-) mice lacking GV sPLA(2) (GV DKO) were infused with 1000 ng/kg per minute Ang II for up to 28 days. Increases in systolic blood pressure, plasma aldosterone level, and urinary and heart prostanoids were similar in apoE(-/-) and GV DKO mice after Ang II infusion. The incidence of aortic rupture in Ang II-infused GV DKO mice (10%) was significantly reduced compared with apoE(-/-) mice (29.4%). Although the incidence of AAA in GV DKO mice (81.3%) and apoE(-/-) mice (100%) was similar, the mean percentage increase in maximal luminal diameter of abdominal aortas was significantly smaller in GV DKO mice (68.5% ± 7.7%) compared with apoE(-/-) mice (92.6% ± 8.3%). Deficiency of GV sPLA(2) resulted in increased Ang II-induced cardiac fibrosis that was most pronounced in perivascular regions. Perivascular collagen, visualized by picrosirius red staining, was associated with increased TUNEL staining and increased immunopositivity for macrophages and myofibroblasts and nicotinamide adenine dinucleotide phosphate oxidase (NOX)-2 and NOX-4, respectively. Our findings indicate that GV sPLA(2) modulates pathological responses to Ang II, with different outcomes for AAA and cardiac fibrosis.
Collapse
Affiliation(s)
- Boris B Boyanovsky
- Endocrinology Division, the Department of Internal Medicine, University of Kentucky, Lexington, USA.
| | | | | | | | | |
Collapse
|
32
|
Differences between group X and group V secretory phospholipase A(2) in lipolytic modification of lipoproteins. Cell Mol Biol Lett 2012; 17:459-78. [PMID: 22706677 PMCID: PMC6275602 DOI: 10.2478/s11658-012-0019-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/31/2012] [Indexed: 11/20/2022] Open
Abstract
Secretory phospholipases A(2) (sPLA(2)s) are a diverse family of low molecular mass enzymes (13-18 kDa) that hydrolyze the sn-2 fatty acid ester bond of glycerophospholipids to produce free fatty acids and lysophospholipids. We have previously shown that group X sPLA(2) (sPLA(2)-X) had a strong hydrolyzing activity toward phosphatidylcholine in low-density lipoprotein (LDL) linked to the formation of lipid droplets in the cytoplasm of macrophages. Here, we show that group V sPLA(2) (sPLA(2)-V) can also cause the lipolysis of LDL, but its action differs remarkably from that of sPLA(2)-X in several respects. Although sPLA(2)-V released almost the same amount of fatty acids from LDL, it released more linoleic acid and less arachidonic acid than sPLA(2)-X. In addition, the requirement of Ca(2+) for the lipolysis of LDL was about 10-fold higher for sPLA(2)-V than sPLA(2)-X. In fact, the release of fatty acids from human serum was hardly detectable upon incubation with sPLA(2)-V in the presence of sodium citrate, which contrasted with the potent response to sPLA(2)-X. Moreover, sPLA(2)-X, but not sPLA(2)-V, was found to specifically interact with LDL among the serum proteins, as assessed by gel-filtration chromatography as well as sandwich enzyme-immunosorbent assay using anti-sPLA(2)-X and anti-apoB antibodies. Surface plasmon resonance studies have revealed that sPLA2-X can bind to LDL with high-affinity (K(d) = 3.1 nM) in the presence of Ca(2+). Selective interaction of sPLA(2)-X with LDL might be involved in the efficient hydrolysis of cell surface or intracellular phospholipids during foam cell formation.
Collapse
|
33
|
Zhan C, Wang J, Kolko M. Diverse Regulation of Retinal Pigment Epithelium Phagocytosis of Photoreceptor Outer Segments by Calcium-Independent Phospholipase A2, Group VIA and Secretory Phospholipase A2, Group IB. Curr Eye Res 2012; 37:930-40. [DOI: 10.3109/02713683.2012.691598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Sergouniotis P, Davidson A, Mackay D, Lenassi E, Li Z, Robson A, Yang X, Kam J, Isaacs T, Holder G, Jeffery G, Beck J, Moore A, Plagnol V, Webster A. Biallelic mutations in PLA2G5, encoding group V phospholipase A2, cause benign fleck retina. Am J Hum Genet 2011; 89:782-91. [PMID: 22137173 DOI: 10.1016/j.ajhg.2011.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/02/2011] [Accepted: 11/07/2011] [Indexed: 10/14/2022] Open
Abstract
Flecked-retina syndromes, including fundus flavimaculatus, fundus albipunctatus, and benign fleck retina, comprise a group of disorders with widespread or limited distribution of yellow-white retinal lesions of various sizes and configurations. Three siblings who have benign fleck retina and were born to consanguineous parents are the basis of this report. A combination of homozygosity mapping and exome sequencing helped to identify a homozygous missense mutation, c.133G>T (p.Gly45Cys), in PLA2G5, a gene encoding a secreted phospholipase (group V phospholipase A(2)). A screen of a further four unrelated individuals with benign fleck retina detected biallelic variants in the same gene in three patients. In contrast, no loss of function or common (minor-allele frequency>0.05%) nonsynonymous PLA2G5 variants have been previously reported (EVS, dbSNP, 1000 Genomes Project) or were detected in an internal database of 224 exomes (from subjects with adult onset neurodegenerative disease and without a diagnosis of ophthalmic disease). All seven affected individuals had fundoscopic features compatible with those previously described in benign fleck retina and no visual or electrophysiological deficits. No medical history of major illness was reported. Levels of low-density lipoprotein were mildly elevated in two patients. Optical coherence tomography and fundus autofluorescence findings suggest that group V phospholipase A(2) plays a role in the phagocytosis of photoreceptor outer-segment discs by the retinal pigment epithelium. Surprisingly, immunohistochemical staining of human retinal tissue revealed localization of the protein predominantly in the inner and outer plexiform layers.
Collapse
|
35
|
Karray A, Ben Ali Y, Boujelben J, Amara S, Carrière F, Gargouri Y, Bezzine S. Drastic changes in the tissue-specific expression of secreted phospholipases A2 in chicken pulmonary disease. Biochimie 2011; 94:451-60. [PMID: 21893157 PMCID: PMC7117035 DOI: 10.1016/j.biochi.2011.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 08/17/2011] [Indexed: 12/16/2022]
Abstract
Infectious bronchitis is one of the most important diseases in poultry and it causes major economic losses. Infectious bronchitis is an acute, highly contagious, viral disease of chickens, characterized by rales, coughing, and sneezing. Because secreted phospholipases A2 (sPLA2) are involved in inflammatory processes, the gene expressions of sPLA2s were investigated in both healthy chickens and chickens with infectious bronchitis and lung inflammation. The draft chicken genome was first scanned using human sPLA2 sequences to identify chicken sPLA2s (ChPLA2), chicken total mRNA were isolated and RT-PCR experiments were performed to amplify and then sequence orthologous cDNAs. Full-length cDNA sequences of ChPLA2-IB, -IIA, -IIE, -V and -X were cloned. The high degree of sequence identity of 50–70% between the avian and mammalian (human and mouse) sPLA2 orthologs suggests a conservation of important enzymatic functions for these phospholipases. Quantitation by qPCR of the transcript levels of ChPLA2-IB, -IIA, -IIE, -V and -X in several tissues from healthy chicken indicated that the expression patterns and mRNA levels diverged among the phospholipases tested. In chicken with infectious bronchitis, an over expression of ChPLA2-V was observed in lungs and spleen in comparison with healthy chicken. These findings suggest that ChPLA2-V could be a potential biomarker for lung inflammation. Conversely, a down regulation of ChPLA2-IB, -IIA and -X was observed in lungs and spleen in case of infectious bronchitis. A significant increase in the expression level of ChPLA2-X and ChPLA2-IB was also noticed in pancreas. No or minor changes have been detected in the expression of ChPLA2-IIE in lungs and small intestine, but it shows a significant increase in several infected tissues.
Collapse
Affiliation(s)
- Aida Karray
- Laboratoire de Biochimie et de Génie Enzymatique des Lipases, ENIS Route de Soukra, université de Sfax, Tunisia
| | | | | | | | | | | | | |
Collapse
|
36
|
Jemel I, Ii H, Oslund RC, Payré C, Dabert-Gay AS, Douguet D, Chargui K, Scarzello S, Gelb MH, Lambeau G. Group X secreted phospholipase A2 proenzyme is matured by a furin-like proprotein convertase and releases arachidonic acid inside of human HEK293 cells. J Biol Chem 2011; 286:36509-21. [PMID: 21878635 DOI: 10.1074/jbc.m111.268540] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Among mammalian secreted phospholipases A(2) (sPLA(2)s), group X sPLA(2) has the most potent hydrolyzing activity toward phosphatidylcholine and is involved in arachidonic acid (AA) release. Group X sPLA(2) is produced as a proenzyme and contains a short propeptide of 11 amino acids ending with a dibasic motif, suggesting cleavage by proprotein convertases. Although the removal of this propeptide is clearly required for enzymatic activity, the cellular location and the protease(s) involved in proenzyme conversion are unknown. Here we have analyzed the maturation of group X sPLA(2) in HEK293 cells, which have been extensively used to analyze sPLA(2)-induced AA release. Using recombinant mouse (PromGX) and human (ProhGX) proenzymes; HEK293 cells transfected with cDNAs coding for full-length ProhGX, PromGX, and propeptide mutants; and various permeable and non-permeable sPLA(2) inhibitors and protease inhibitors, we demonstrate that group X sPLA(2) is mainly converted intracellularly and releases AA before externalization from the cell. Most strikingly, the exogenous proenzyme does not elicit AA release, whereas the transfected proenzyme does elicit AA release in a way insensitive to non-permeable sPLA(2) inhibitors. In transfected cells, a permeable proprotein convertase inhibitor, but not a non-permeable one, prevents group X sPLA(2) maturation and partially blocks AA release. Mutations at the dibasic motif of the propeptide indicate that the last basic residue is required and sufficient for efficient maturation and AA release. All together, these results argue for the intracellular maturation of group X proenzyme in HEK293 cells by a furin-like proprotein convertase, leading to intracellular release of AA during secretion.
Collapse
Affiliation(s)
- Ikram Jemel
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, CNRS et Université de Nice-Sophia-Antipolis, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
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: 42] [Impact Index Per Article: 3.0] [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
|
38
|
Yamamoto K, Isogai Y, Sato H, Taketomi Y, Murakami M. Secreted phospholipase A2, lipoprotein hydrolysis, and atherosclerosis: integration with lipidomics. Anal Bioanal Chem 2011; 400:1829-42. [PMID: 21445663 PMCID: PMC3098357 DOI: 10.1007/s00216-011-4864-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Revised: 02/14/2011] [Accepted: 03/01/2011] [Indexed: 01/22/2023]
Abstract
Phospholipase A2 (PLA2) is a group of enzymes that hydrolyze the sn-2 position of glycerophospholipids to yield fatty acids and lysophospholipids. Of many PLA2s or related enzymes identified to date, secreted PLA2s (sPLA2s) comprise the largest family that contains 10 catalytically active isozymes. Besides arachidonic acid released from cellular membranes for eicosanoid synthesis, several if not all sPLA2s have recently been implicated in hydrolysis of phospholipids in lipoprotein particles. The sPLA2-processed low-density lipoprotein (LDL) particles contain a large amount of lysophospholipids and exhibit the property of “small-dense” or “modified” LDL, which facilitates foam cell formation from macrophages. Transgenic overexpression of these sPLA2s leads to development of atherosclerosis in mice. More importantly, genetic deletion or pharmacological inhibition of particular sPLA2s significantly attenuates atherosclerosis and aneurysm. In this article, we will give an overview of current understanding of the role of sPLA2s in atherosclerosis, with recent lipidomics data showing the action of a subset of sPLA2s on lipoprotein phospholipids.
Collapse
Affiliation(s)
- Kei Yamamoto
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | | | | | | | | |
Collapse
|
39
|
Murakami M, Sato H, Taketomi Y, Yamamoto K. Integrated lipidomics in the secreted phospholipase A(2) biology. Int J Mol Sci 2011; 12:1474-95. [PMID: 21673902 PMCID: PMC3111613 DOI: 10.3390/ijms12031474] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 02/18/2011] [Accepted: 02/24/2011] [Indexed: 12/22/2022] Open
Abstract
Mammalian genomes encode genes for more than 30 phospholipase A(2)s (PLA(2)s) or related enzymes, which are subdivided into several subgroups based on their structures, catalytic mechanisms, localizations and evolutionary relationships. More than one third of the PLA(2) enzymes belong to the secreted PLA(2) (sPLA(2)) family, which consists of low-molecular-weight, Ca(2+)-requiring extracellular enzymes, with a His-Asp catalytic dyad. Individual sPLA(2) isoforms exhibit unique tissue and cellular localizations and enzymatic properties, suggesting their distinct pathophysiological roles. Recent studies using transgenic and knockout mice for several sPLA(2) isoforms, in combination with lipidomics approaches, have revealed their distinct contributions to various biological events. Herein, we will describe several examples of sPLA(2)-mediated phospholipid metabolism in vivo, as revealed by integrated analysis of sPLA(2) transgenic/knockout mice and lipid mass spectrometry. Knowledge obtained from this approach greatly contributes to expanding our understanding of the sPLA(2) biology and pathophysiology.
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; E-Mails: (H.S.); (Y.T.); and (K.Y.)
| | - Hiroyasu Sato
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; E-Mails: (H.S.); (Y.T.); and (K.Y.)
| | - Yoshitaka Taketomi
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; E-Mails: (H.S.); (Y.T.); and (K.Y.)
| | - Kei Yamamoto
- Lipid Metabolism Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; E-Mails: (H.S.); (Y.T.); and (K.Y.)
| |
Collapse
|
40
|
Yamamoto K, Taketomi Y, Isogai Y, Miki Y, Sato H, Masuda S, Nishito Y, Morioka K, Ishimoto Y, Suzuki N, Yokota Y, Hanasaki K, Ishikawa Y, Ishii T, Kobayashi T, Fukami K, Ikeda K, Nakanishi H, Taguchi R, Murakami M. Hair follicular expression and function of group X secreted phospholipase A2 in mouse skin. J Biol Chem 2011; 286:11616-31. [PMID: 21266583 DOI: 10.1074/jbc.m110.206714] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although perturbed lipid metabolism can often lead to skin abnormality, the role of phospholipase A(2) (PLA(2)) in skin homeostasis is poorly understood. In the present study we found that group X-secreted PLA(2) (sPLA(2)-X) was expressed in the outermost epithelium of hair follicles in synchrony with the anagen phase of hair cycling. Transgenic mice overexpressing sPLA(2)-X (PLA2G10-Tg) displayed alopecia, which was accompanied by hair follicle distortion with reduced expression of genes related to hair development, during a postnatal hair cycle. Additionally, the epidermis and sebaceous glands of PLA2G10-Tg skin were hyperplasic. Proteolytic activation of sPLA(2)-X in PLA2G10-Tg skin was accompanied by preferential hydrolysis of phosphatidylethanolamine species with polyunsaturated fatty acids as well as elevated production of some if not all eicosanoids. Importantly, the skin of Pla2g10-deficient mice had abnormal hair follicles with noticeable reduction in a subset of hair genes, a hypoplasic outer root sheath, a reduced number of melanin granules, and unexpected up-regulation of prostanoid synthesis. Collectively, our study highlights the spatiotemporal expression of sPLA(2)-X in hair follicles, the presence of skin-specific machinery leading to sPLA(2)-X activation, a functional link of sPLA(2)-X with hair follicle homeostasis, and compartmentalization of the prostanoid pathway in hair follicles and epidermis.
Collapse
Affiliation(s)
- Kei Yamamoto
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
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: 389] [Impact Index Per Article: 25.9] [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
|
42
|
Mallat Z, Lambeau G, Tedgui A. Lipoprotein-associated and secreted phospholipases A₂ in cardiovascular disease: roles as biological effectors and biomarkers. Circulation 2010; 122:2183-200. [PMID: 21098459 DOI: 10.1161/circulationaha.110.936393] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Ziad Mallat
- INSERM, Paris-Cardiovascular Research Center, Université Paris Descartes, UMR, Paris, France
| | | | | |
Collapse
|
43
|
Shridas P, Bailey WM, Gizard F, Oslund RC, Gelb MH, Bruemmer D, Webb NR. Group X secretory phospholipase A2 negatively regulates ABCA1 and ABCG1 expression and cholesterol efflux in macrophages. Arterioscler Thromb Vasc Biol 2010; 30:2014-21. [PMID: 20844270 DOI: 10.1161/atvbaha.110.210237] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE GX sPLA(2) potently hydrolyzes plasma membranes to generate lysophospholipids and free fatty acids; it has been implicated in inflammatory diseases, including atherosclerosis. To identify a novel role for group X (GX) secretory phospholipase A(2) (sPLA(2)) in modulating ATP binding casette transporter A1 (ABCA1) and ATP binding casette transporter G1 (ABCG1) expression and, therefore, macrophage cholesterol efflux. METHODS AND RESULTS The overexpression or exogenous addition of GX sPLA(2) significantly reduced ABCA1 and ABCG1 expression in J774 macrophage-like cells, whereas GX sPLA(2) deficiency in mouse peritoneal macrophages was associated with enhanced expression. Altered ABC transporter expression led to reduced cholesterol efflux in GX sPLA(2)-overexpressing J774 cells and increased efflux in GX sPLA(2)-deficient mouse peritoneal macrophages. Gene regulation was dependent on GX sPLA(2) catalytic activity, mimicked by arachidonic acid and abrogated when liver X receptor (LXR)α/β expression was suppressed, and partially reversed by the LXR agonist T0901317. Reporter assays indicated that GX sPLA(2) suppresses the ability of LXR to transactivate its promoters through a mechanism involving the C-terminal portion of LXR spanning the ligand-binding domain. CONCLUSIONS GX sPLA(2) modulates gene expression in macrophages by generating lipolytic products that suppress LXR activation. GX sPLA(2) may play a previously unrecognized role in atherosclerotic lipid accumulation by negatively regulating the genes critical for cellular cholesterol efflux.
Collapse
Affiliation(s)
- Preetha Shridas
- Graduate Center for Nutritional Sciences, Saha Cardiovascular Research Center, University of Kentucky Medical Center, Lexington 40536-0200, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Escoffier J, Jemel I, Tanemoto A, Taketomi Y, Payre C, Coatrieux C, Sato H, Yamamoto K, Masuda S, Pernet-Gallay K, Pierre V, Hara S, Murakami M, De Waard M, Lambeau G, Arnoult C. Group X phospholipase A2 is released during sperm acrosome reaction and controls fertility outcome in mice. J Clin Invest 2010; 120:1415-28. [PMID: 20424324 DOI: 10.1172/jci40494] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 02/10/2010] [Indexed: 11/17/2022] Open
Abstract
Ejaculated mammalian sperm must undergo a maturation process called capacitation before they are able to fertilize an egg. Several studies have suggested a role for members of the secreted phospholipase A2 (sPLA2) family in capacitation, acrosome reaction (AR), and fertilization, but the molecular nature of these enzymes and their specific roles have remained elusive. Here, we have demonstrated that mouse group X sPLA2 (mGX) is the major enzyme present in the acrosome of spermatozoa and that it is released in an active form during capacitation through spontaneous AR. mGX-deficient male mice produced smaller litters than wild-type male siblings when crossed with mGX-deficient females. Further analysis revealed that spermatozoa from mGX-deficient mice exhibited lower rates of spontaneous AR and that this was associated with decreased in vitro fertilization (IVF) efficiency due to a drop in the fertilization potential of the sperm and an increased rate of aborted embryos. Treatment of sperm with sPLA2 inhibitors and antibodies specific for mGX blocked spontaneous AR of wild-type sperm and reduced IVF success. Addition of lysophosphatidylcholine, a catalytic product of mGX, overcame these deficiencies. Finally, recombinant mGX triggered AR and improved IVF outcome. Taken together, our results highlight a paracrine role for mGX during capacitation in which the enzyme primes sperm for efficient fertilization and boosts premature AR of a likely phospholipid-damaged sperm subpopulation to eliminate suboptimal sperm from the pool available for fertilization.
Collapse
|
46
|
Shridas P, Bailey WM, Boyanovsky BB, Oslund RC, Gelb MH, Webb NR. Group X secretory phospholipase A2 regulates the expression of steroidogenic acute regulatory protein (StAR) in mouse adrenal glands. J Biol Chem 2010; 285:20031-9. [PMID: 20421306 DOI: 10.1074/jbc.m109.090423] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We developed C57BL/6 mice with targeted deletion of group X secretory phospholipase A(2) (GX KO). These mice have approximately 80% higher plasma corticosterone concentrations compared with wild-type (WT) mice under both basal and adrenocorticotropic hormone (ACTH)-induced stress conditions. This increased corticosterone level was not associated with increased circulating ACTH or a defect in the hypothalamic-pituitary axis as evidenced by a normal response to dexamethasone challenge. Primary cultures of adrenal cells from GX KO mice exhibited significantly increased corticosteroid secretion compared with WT cells. Conversely, overexpression of GX secretory phospholipase A(2) (sPLA(2)), but not a catalytically inactive mutant form of GX sPLA(2), significantly reduced steroid production 30-40% in Y1 mouse adrenal cell line. This effect was reversed by the sPLA(2) inhibitor, indoxam. Silencing of endogenous M-type receptor expression did not restore steroid production in GX sPLA(2)-overexpressing Y1 cells, ruling out a role for this sPLA(2) receptor in this regulatory process. Expression of steroidogenic acute regulatory protein (StAR), the rate-limiting protein in corticosteroid production, was approximately 2-fold higher in adrenal glands of GX KO mice compared with WT mice, whereas StAR expression was suppressed in Y1 cells overexpressing GX sPLA(2). Results from StAR-promoter luciferase reporter gene assays indicated that GX sPLA(2) antagonizes StAR promoter activity and liver X receptor-mediated StAR promoter activation. In summary, GX sPLA(2) is expressed in mouse adrenal glands and functions to negatively regulate corticosteroid synthesis, most likely by negatively regulating StAR expression.
Collapse
Affiliation(s)
- Preetha Shridas
- Graduate Center for Nutritional Sciences, the Cardiovascular Research Center, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA
| | | | | | | | | | | |
Collapse
|
47
|
Murakami M, Taketomi Y, Girard C, Yamamoto K, Lambeau G. Emerging roles of secreted phospholipase A2 enzymes: Lessons from transgenic and knockout mice. Biochimie 2010; 92:561-82. [PMID: 20347923 DOI: 10.1016/j.biochi.2010.03.015] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 03/18/2010] [Indexed: 11/15/2022]
Abstract
Among the emerging phospholipase A(2) (PLA(2)) superfamily, the secreted PLA(2) (sPLA(2)) family consists of low-molecular-mass, Ca(2+)-requiring extracellular enzymes with a His-Asp catalytic dyad. To date, more than 10 sPLA(2) enzymes have been identified in mammals. Individual sPLA(2)s exhibit unique tissue and cellular localizations and enzymatic properties, suggesting their distinct pathophysiological roles. Despite numerous enzymatic and cell biological studies on this enzyme family in the past two decades, their precise in vivo functions still remain largely obscure. Recent studies using transgenic and knockout mice for several sPLA(2) enzymes, in combination with lipidomics approaches, have opened new insights into their distinct contributions to various biological events such as food digestion, host defense, inflammation, asthma and atherosclerosis. In this article, we overview the latest understanding of the pathophysiological functions of individual sPLA(2) isoforms fueled by studies employing transgenic and knockout mice for several sPLA(2)s.
Collapse
Affiliation(s)
- Makoto Murakami
- Biomembrane Signaling Project, The Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | | | | | | | | |
Collapse
|
48
|
Goracci G, Ferrini M, Nardicchi V. Low Molecular Weight Phospholipases A2 in Mammalian Brain and Neural Cells: Roles in Functions and Dysfunctions. Mol Neurobiol 2010; 41:274-89. [DOI: 10.1007/s12035-010-8108-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 02/11/2010] [Indexed: 12/14/2022]
|
49
|
Sun GY, Shelat PB, Jensen MB, He Y, Sun AY, Simonyi A. Phospholipases A2 and inflammatory responses in the central nervous system. Neuromolecular Med 2009; 12:133-48. [PMID: 19855947 DOI: 10.1007/s12017-009-8092-z] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 09/25/2009] [Indexed: 12/21/2022]
Abstract
Phospholipases A2 (PLA2s) belong to a superfamily of enzymes responsible for hydrolyzing the sn-2 fatty acids of membrane phospholipids. These enzymes are known to play multiple roles for maintenance of membrane phospholipid homeostasis and for production of a variety of lipid mediators. Over 20 different types of PLA2s are present in the mammalian cells, and in snake and bee venom. Despite their common function in hydrolyzing fatty acids of phospholipids, they are diversely encoded by a number of genes and express proteins that are regulated by different mechanisms. Recent studies have focused on the group IV calcium-dependent cytosolic cPLA2, the group VI calcium-independent iPLA2, and the group II small molecule secretory sPLA2. In the central nervous system (CNS), these PLA2s are distributed among neurons and glial cells. Although the physiological role of these PLA2s in regulating neural cell function has not yet been clearly elucidated, there is increasing evidence for their involvement in receptor signaling and transcriptional pathways that link oxidative events to inflammatory responses that underline many neurodegenerative diseases. Recent studies also reveal an important role of cPLA2 in modulating neuronal excitatory functions, sPLA2 in the inflammatory responses, and iPLA2 with childhood neurologic disorders associated with brain iron accumulation. The goal for this review is to better understand the structure and function of these PLA2s and to highlight specific types of PLA2s and their cross-talk mechanisms in these inflammatory responses under physiological and pathological conditions in the CNS.
Collapse
Affiliation(s)
- Grace Y Sun
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO 65211, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Fonseca RG, Ferreira TL, Ward RJ. Refolding and purification of the human secreted group IID phospholipase A2 expressed as inclusion bodies in Escherichia coli. Protein Expr Purif 2009; 67:82-7. [DOI: 10.1016/j.pep.2009.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 04/03/2009] [Accepted: 04/09/2009] [Indexed: 11/29/2022]
|