1
|
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: 29] [Impact Index Per Article: 14.5] [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
|
2
|
Shi D, Feng C, Xie J, Zhang X, Dai H, Yan L. Recent Progress of Nanomedicine on Secreted Phospholipase A2 as a Potential Therapeutic Target. J Mater Chem B 2022; 10:7349-7360. [DOI: 10.1039/d2tb00608a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Overexpressed secretory phospholipase A2 (sPLA2) is found in many inflammatory diseases and various types of cancer. sPLA2 can catalyze the hydrolysis of phospholipid sn-2 ester bond to lysophosphatidylcholine and free...
Collapse
|
3
|
Smith JW, Barlas RS, Mamas MA, Boekholdt SM, Mallat Z, Luben RN, Wareham NJ, Khaw KT, Myint PK. Association between serum secretory phospholipase A2 and risk of ischaemic stroke. Eur J Neurol 2021; 28:3650-3655. [PMID: 34216520 DOI: 10.1111/ene.15004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Previous literature has demonstrated an association between high serum levels of type II secretory phospholipase A2 (sPLA2) concentration and an increased risk of coronary artery disease. However, such association has not been established in terms of ischaemic stroke risk. The aim was to evaluate the association between both sPLA2 concentration and activity as continuous variables with risk of future ischaemic stroke. METHODS A nested case-control study was conducted using data from the European Prospective Investigation into Cancer-Norfolk study. Cases (n = 145) in the current study were participants who developed ischaemic stroke during follow-up, with controls (n = 290) matched in a 2:1 ratio based on age and sex. Statistical analyses were performed using SPSS (version 25.0) software. Logistic regression was used to determine odds ratios (OR) and corresponding 95% confidence intervals (95% CIs) for ischaemic stroke. RESULTS After adjusting for a wide array of cardiovascular confounders, sPLA2 activity was found to be associated with an increased risk of ischaemic stroke using both multiple imputations with chained equations and complete case analysis: OR 1.20 (95% CI 1.01-1.43) and OR 1.23 (95% CI 1.01-1.49), respectively. However, sPLA2 concentration was not found to be associated with increased risk of ischaemic stroke. CONCLUSIONS The activity of sPLA2, but not sPLA2 concentration, is associated with an increased risk of future ischaemic stroke. This finding may be significant in risk group stratification, allowing targeted prophylactic treatment, or the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Jed W Smith
- Ageing Clinical and Experimental Research, Institute of Applied Health Sciences, Aberdeen, UK
| | - Raphae S Barlas
- Ageing Clinical and Experimental Research, Institute of Applied Health Sciences, Aberdeen, UK
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Institute for Primary Care and Health Sciences, Stoke-on-Trent, UK
| | - S Matthijs Boekholdt
- Department of Cardiology and Vascular Medicine (M.B.), Academic Medical Center, Amsterdam, The Netherlands
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Robert N Luben
- Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Phyo K Myint
- Ageing Clinical and Experimental Research, Institute of Applied Health Sciences, Aberdeen, UK
| |
Collapse
|
4
|
Morris G, Berk M, Walder K, O'Neil A, Maes M, Puri BK. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci Biobehav Rev 2021; 128:35-57. [PMID: 34118292 DOI: 10.1016/j.neubiorev.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic systemic inflammation is associated with an increased risk of cardiovascular disease in an environment of low low-density lipoprotein (LDL) and low total cholesterol and with the pathophysiology of neuroprogressive disorders. The causes and consequences of this lipid paradox are explored. Circulating activated neutrophils can release inflammatory molecules such as myeloperoxidase and the pro-inflammatory cytokines interleukin-1 beta, interleukin-6 and tumour necrosis factor-alpha. Since activated neutrophils are associated with atherosclerosis and cardiovascular disease and with major depressive disorder, bipolar disorder and schizophrenia, it seems reasonable to hypothesise that the inflammatory molecules released by them may act as mediators of the link between systemic inflammation and the development of atherosclerosis in neuroprogressive disorders. This hypothesis is tested by considering the association at a molecular level of systemic inflammation with increased LDL oxidation; increased small dense LDL levels; increased lipoprotein (a) concentration; secretory phospholipase A2 activation; cytosolic phospholipase A2 activation; increased platelet activation; decreased apolipoprotein A1 levels and function; decreased paroxonase-1 activity; hyperhomocysteinaemia; and metabolic endotoxaemia. These molecular mechanisms suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
| | | |
Collapse
|
5
|
Lysophosphatidylcholine in phospholipase A 2-modified LDL triggers secretion of angiopoietin 2. Atherosclerosis 2021; 327:87-99. [PMID: 34020784 DOI: 10.1016/j.atherosclerosis.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Secretory phospholipase A2 (PLA2) hydrolyzes LDL phospholipids generating modified LDL particles (PLA2-LDL) with increased atherogenic properties. Exocytosis of Weibel-Palade bodies (WPB) releases angiopoietin 2 (Ang2) and externalizes P-selectin, which both play important roles in vascular inflammation. Here, we investigated the effects of PLA2-LDL on exocytosis of WPBs. METHODS Human coronary artery endothelial cells (HCAECs) were stimulated with PLA2- LDL, and its uptake and effect on Ang2 release, leukocyte adhesion, and intracellular calcium levels were measured. The effects of PLA2-LDL on Ang2 release and WPB exocytosis were measured in and ex vivo in mice. RESULTS Exposure of HCAECs to PLA2-LDL triggered Ang2 secretion and promoted leukocyte-HCAEC interaction. Lysophosphatidylcholine was identified as a critical component of PLA2-LDL regulating the WPB exocytosis, which was mediated by cell-surface proteoglycans, phospholipase C, intracellular calcium, and cytoskeletal remodeling. PLA2-LDL also induced murine endothelial WPB exocytosis in blood vessels in and ex vivo, as evidenced by secretion of Ang2 in vivo, P-selectin translocation to plasma membrane in intact endothelial cells in thoracic artery and tracheal vessels, and reduced Ang2 staining in tracheal endothelial cells. Finally, in contrast to normal human coronary arteries, in which Ang2 was present only in the endothelial layer, at sites of advanced atherosclerotic lesions, Ang2 was detected also in the intima, media, and adventitia. CONCLUSIONS Our studies reveal PLA2-LDL as a potent agonist of endothelial WPB exocytosis, resulting in increased secretion of Ang2 and translocation of P-selectin. The results provide mechanistic insight into PLA2-LDL-dependent promotion of vascular inflammation and atherosclerosis.
Collapse
|
6
|
Sanda GM, Stancu CS, Deleanu M, Toma L, Niculescu LS, Sima AV. Aggregated LDL turn human macrophages into foam cells and induce mitochondrial dysfunction without triggering oxidative or endoplasmic reticulum stress. PLoS One 2021; 16:e0245797. [PMID: 33493198 PMCID: PMC7833132 DOI: 10.1371/journal.pone.0245797] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/07/2021] [Indexed: 11/18/2022] Open
Abstract
Uptake of modified lipoproteins by macrophages turns them into foam cells, the hallmark of the atherosclerotic plaque. The initiation and progression of atherosclerosis have been associated with mitochondrial dysfunction. It is known that aggregated low-density lipoproteins (agLDL) induce massive cholesterol accumulation in macrophages in contrast with native LDL (nLDL) and oxidized LDL (oxLDL). In the present study we aimed to assess the effect of agLDL on the mitochondria and ER function in macrophage-derived foam cells, in an attempt to estimate the potential of these cells, known constituents of early fatty streaks, to generate atheroma in the absence of oxidative stress. Results show that agLDL induce excessive accumulation of free (FC) and esterified cholesterol in THP-1 macrophages and determine mitochondrial dysfunction expressed as decreased mitochondrial membrane potential and diminished intracellular ATP levels, without generating mitochondrial reactive oxygen species (ROS) production. AgLDL did not stimulate intracellular ROS (superoxide anion or hydrogen peroxide) production, and did not trigger endoplasmic reticulum stress (ERS) or apoptosis. In contrast to agLDL, oxLDL did not modify FC levels, but stimulated the accumulation of 7-ketocholesterol in the cells, generating oxidative stress which is associated with an increased mitochondrial dysfunction, ERS and apoptosis. Taken together, our results reveal that agLDL induce foam cells formation and mild mitochondrial dysfunction in human macrophages without triggering oxidative or ERS. These data could partially explain the early formation of fatty streaks in the intima of human arteries by interaction of monocyte-derived macrophages with non-oxidatively aggregated LDL generating foam cells, which cannot evolve into atherosclerotic plaques in the absence of the oxidative stress.
Collapse
Affiliation(s)
- Gabriela M Sanda
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Camelia S Stancu
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Mariana Deleanu
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.,Faculty of Biotechnology, University of Agronomical Sciences and Veterinary Medicine, Bucharest, Romania
| | - Laura Toma
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Loredan S Niculescu
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Anca V Sima
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
7
|
Fras Z, Tršan J, Banach M. On the present and future role of Lp-PLA 2 in atherosclerosis-related cardiovascular risk prediction and management. Arch Med Sci 2021; 17:954-964. [PMID: 34336025 PMCID: PMC8314407 DOI: 10.5114/aoms.2020.98195] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/23/2022] Open
Abstract
Circulating concentration and activity of secretory phospholipase A2 (sPLA2) and lipoprotein-associated phospholipase A2 (Lp-PLA2) have been proven as biomarkers of increased risk of atherosclerosis-related cardiovascular disease (ASCVD). Lp-PLA2 might be part of the atherosclerotic process and may contribute to plaque destabilisation through inflammatory activity within atherosclerotic lesions. However, all attempts to translate the inhibition of phospholipase into clinically beneficial ASCVD risk reduction, including in randomised studies, by either non-specific inhibition of sPLA2 (by varespladib) or specific Lp-PLA2 inhibition by darapladib, unexpectedly failed. This gives us a strong imperative to continue research aimed at a better understanding of how Lp-PLA2 and sPLA2 regulate vascular inflammation and atherosclerotic plaque development. From the clinical viewpoint there is a need to establish and validate the existing and emerging novel anti-inflammatory therapeutic strategies to fight against ASCVD development, by using potentially better animal models and differently designed clinical trials in humans.
Collapse
Affiliation(s)
- Zlatko Fras
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Chair of Internal Medicine, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Tršan
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| |
Collapse
|
8
|
Group IIA Secretory Phospholipase A2 Predicts Graft Failure and Mortality in Renal Transplant Recipients by Mediating Decreased Kidney Function. J Clin Med 2020; 9:jcm9051282. [PMID: 32365505 PMCID: PMC7288094 DOI: 10.3390/jcm9051282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 01/14/2023] Open
Abstract
The acute phase protein group IIA secretory phospholipase A2 (sPLA2-IIA) has intrinsic proatherosclerotic properties. The present prospective cohort study investigated whether plasma sPLA2-IIA associates with graft failure, cardiovascular, and all-cause mortality in renal transplant recipients (RTRs), patients with accelerated atherosclerosis formation both systemically and within the graft. In 511 RTRs from a single academic center with stable graft function >1 year, baseline plasma sPLA2-IIA was determined by ELISA. Primary end points were death-censored graft failure and mortality (median follow-up, 7.0 years). Baseline sPLA2-IIA was higher in RTRs than in healthy controls (median 384 ng/dL (range 86–6951) vs. 185 ng/dL (range 104–271), p < 0.001). Kaplan–Meier analysis demonstrated increased risk for graft failure (p = 0.002), as well as cardiovascular (p < 0.001) and all-cause mortality (p < 0.001), with increasing sPLA2-IIA quartiles. Cox regression showed strong associations of sPLA2-IIA with increased risks of graft failure (hazard ratio (HR) = 1.42 (1.11–1.83), p = 0.006), as well as cardiovascular (HR = 1.48 (1.18−1.85), p = 0.001) and all-cause mortality (HR = 1.39 (1.17−1.64), p < 0.001), dependent on parameters of kidney function. Renal function during follow-up declined faster in RTRs with higher baseline sPLA2-IIA levels. In RTRs, sPLA2-IIA is a significant predictive biomarker for chronic graft failure, as well as overall and cardiovascular disease mortality dependent on kidney function. This dependency is conceivably explained by sPLA2-IIA impacting negatively on kidney function.
Collapse
|
9
|
Benitez-Amaro A, Pallara C, Nasarre L, Rivas-Urbina A, Benitez S, Vea A, Bornachea O, de Gonzalo-Calvo D, Serra-Mir G, Villegas S, Prades R, Sanchez-Quesada JL, Chiva C, Sabido E, Tarragó T, Llorente-Cortés V. Molecular basis for the protective effects of low-density lipoprotein receptor-related protein 1 (LRP1)-derived peptides against LDL aggregation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1302-1316. [PMID: 31077676 DOI: 10.1016/j.bbamem.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/09/2019] [Accepted: 05/01/2019] [Indexed: 01/02/2023]
Abstract
Aggregated LDL is the first ligand reported to interact with the cluster II CR9 domain of low-density lipoprotein receptor-related protein 1 (LRP1). In particular, the C-terminal half of domain CR9, comprising the region Gly1127-Cys1140 exclusively recognizes aggregated LDL and it is crucial for aggregated LDL binding. Our aim was to study the effect of the sequence Gly1127-Cys1140 (named peptide LP3 and its retro-enantio version, named peptide DP3) on the structural characteristics of sphingomyelinase- (SMase) and phospholipase 2 (PLA2)-modified LDL particles. Turbidimetry, gel filtration chromatography (GFC) and transmission electronic microscopy (TEM) analysis showed that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation. Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose gel electrophoresis and high-performance thin-layer chromatography (HPTLC) indicated that LP3 and DP3 prevented SMase-induced alterations in LDL particle size, electric charge and phospholipid content, respectively, but not those induced by PLA2. Western blot analysis showed that LP3 and DP3 counteracted changes in ApoB-100 conformation induced by the two enzymes. LDL proteomics (LDL trypsin digestion followed by mass spectroscopy) and computational modeling methods evidenced that peptides preserve ApoB-100 conformation due to their electrostatic interactions with a basic region of ApoB-100. These results demonstrate that LRP1-derived peptides are protective against LDL aggregation, even in conditions of extreme lipolysis, through their capacity to bind to ApoB-100 regions critical for ApoB-100 conformational preservation. These results suggests that these LRP1(CR9) derived peptides could be promising tools to prevent LDL aggregation induced by the main proteolytic enzymes acting in the arterial intima.
Collapse
Affiliation(s)
- Aleyda Benitez-Amaro
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
| | - Chiara Pallara
- Iproteos S.L., Barcelona Science Park (PCB), Barcelona, Spain
| | - Laura Nasarre
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Andrea Rivas-Urbina
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Angela Vea
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Olga Bornachea
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
| | - David de Gonzalo-Calvo
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain; CIBER Enfermedades Cardiovasculares (CIBERcv), Spain
| | - Gabriel Serra-Mir
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Roger Prades
- Iproteos S.L., Barcelona Science Park (PCB), Barcelona, Spain
| | - José Luís Sanchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBER DIABETES y Enfermedades Metabólicas Asociadas (CIBERdem), Spain
| | - Cristina Chiva
- Proteomics Unit, Centre de Regulació Genòmica, Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabido
- Proteomics Unit, Centre de Regulació Genòmica, Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Teresa Tarragó
- Iproteos S.L., Barcelona Science Park (PCB), Barcelona, Spain
| | - Vicenta Llorente-Cortés
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain; CIBER Enfermedades Cardiovasculares (CIBERcv), Spain.
| |
Collapse
|
10
|
Ligi D, Benitez S, Croce L, Rivas-Urbina A, Puig N, Ordóñez-Llanos J, Mannello F, Sanchez-Quesada JL. Electronegative LDL induces MMP-9 and TIMP-1 release in monocytes through CD14 activation: Inhibitory effect of glycosaminoglycan sulodexide. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3559-3567. [PMID: 30254012 DOI: 10.1016/j.bbadis.2018.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/03/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Electronegative LDL (LDL(-)) is involved in atherosclerosis through the activation of the TLR4/CD14 inflammatory pathway in monocytes. Matrix metalloproteinases (MMP) and their inhibitors (tissue inhibitors of metalloproteinase [TIMP]) are also crucially involved in atherosclerosis, but their modulation by LDL(-) has never been investigated. The aim of this study was to examine the ability of LDL(-) to release MMPs and TIMPs in human monocytes and to determine whether sulodexide (SDX), a glycosaminoglycan-based drug, was able to affect their secretion. APPROACH AND RESULTS Native LDL (LDL(+)) and LDL(-) separated by anion-exchange chromatography were added to THP1-CD14 monocytes in the presence or absence of SDX for 24 h. A panel of 9 MMPs and 4 TIMPs was analyzed in cell supernatants with multiplex immunoassays. The gelatinolytic activity of MMP-9 was assessed by gelatin zymography. LDL(-) stimulated the release of MMP-9 (13-fold) and TIMP-1 (4-fold) in THP1-CD14 monocytes, as well as the gelatinolytic activity of MMP-9. Co-incubation of monocytes with LDL(-) and SDX for 24 h significantly reduced both the release of MMP-9 and TIMP-1 and gelatinase activity. In THP1 cells not expressing CD14, no effect of LDL(-) on MMP-9 or TIMP-1 release was observed. The uptake of DiI-labeled LDL(-) was higher than that of DiI-LDL(+) in THP1-CD14 but not in THP1 cells. This increase was inhibited by SDX. Experiments in microtiter wells coated with SDX demonstrated a specific interaction of LDL(-) with SDX. CONCLUSIONS LDL(-) induced the release of MMP-9 and TIMP-1 in monocytes through CD14. SDX affects the ability of LDL(-) to promote TIMP-1 and MMP-9 release by its interaction with LDL(-).
Collapse
Affiliation(s)
- Daniela Ligi
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy
| | - Sonia Benitez
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Lidia Croce
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy
| | - Andrea Rivas-Urbina
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Núria Puig
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Jordi Ordóñez-Llanos
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; Molecular Biology and Biochemistry Department, Universitat Autònoma de Barcelona (UAB). Cerdanyola del Vallès, Spain
| | - Ferdinando Mannello
- Department of Biomolecular Sciences, Section of Clinical Biochemistry and Molecular Genetics, University Carlo Bo Urbino, Italy.
| | - Jose Luis Sanchez-Quesada
- Cardiovascular Biochemistry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain, C/Sant Antoni M. Claret 167, 08025 Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM).
| |
Collapse
|
11
|
Effects of enzymes on elastic modulus of low-density lipoproteins were investigated using atomic force microscopy. Biochem Biophys Res Commun 2018; 501:607-611. [PMID: 29709480 DOI: 10.1016/j.bbrc.2018.04.211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 11/23/2022]
Abstract
Oxidation of low-density lipoproteins (LDLs) induces development of cardiovascular disease. Recently, reports of studies using atomic force microscopy (AFM) have described that the elastic modulus of metal-induced oxidized LDLs is lower than the modulus before oxidation. However, the mechanisms of change of the elastic modulus have not been well investigated. We postulated that disorder of the LDL structure might decrease the elastic modulus. This study measured the elastic modulus of LDLs before and after enzyme treatment with V8 protease, α-chymotrypsin, and phospholipase A2. After LDLs were obtained from serum by ultracentrifugation, LDLs or enzyme-treated LDLs were physically absorbed. They were crowded on a mica surface. Although V8 protease and α-chymotrypsin did not induce the elastic modulus change, treatment with PLA2 decreased the elastic modulus. The LDL particle size did not change during the enzyme treatment. Results suggest that disordering of the lipid structure of the LDL might contribute to the elastic modulus change. Results show that AFM might be a useful tool to evaluate disorders of complex nanoscale particle structures from lipids and proteins such as lipoproteins.
Collapse
|
12
|
Maaninka K, Nguyen SD, Mäyränpää MI, Plihtari R, Rajamäki K, Lindsberg PJ, Kovanen PT, Öörni K. Human mast cell neutral proteases generate modified LDL particles with increased proteoglycan binding. Atherosclerosis 2018; 275:390-399. [PMID: 29703634 DOI: 10.1016/j.atherosclerosis.2018.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/06/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS Subendothelial interaction of LDL with extracellular matrix drives atherogenesis. This interaction can be strengthened by proteolytic modification of LDL. Mast cells (MCs) are present in atherosclerotic lesions, and upon activation, they degranulate and release a variety of neutral proteases. Here we studied the ability of MC proteases to cleave apoB-100 of LDL and affect the binding of LDL to proteoglycans. METHODS Mature human MCs were differentiated from human peripheral blood-derived CD34+ progenitors in vitro and activated with calcium ionophore to generate MC-conditioned medium. LDL was incubated in the MC-conditioned medium or with individual MC proteases, and the binding of native and modified LDL to isolated human aortic proteoglycans or to human atherosclerotic plaques ex vivo was determined. MC proteases in atherosclerotic human coronary artery lesions were detected by immunofluorescence and qPCR. RESULTS Activated human MCs released the neutral proteases tryptase, chymase, carboxypeptidase A3, cathepsin G, and granzyme B. Of these, cathepsin G degraded most efficiently apoB-100, induced LDL fusion, and enhanced binding of LDL to isolated human aortic proteoglycans and human atherosclerotic lesions ex vivo. Double immunofluoresence staining of human atherosclerotic coronary arteries for tryptase and cathepsin G indicated that lesional MCs contain cathepsin G. In the lesions, expression of cathepsin G correlated with the expression of tryptase and chymase, but not with that of neutrophil proteinase 3. CONCLUSIONS The present study suggests that cathepsin G in human atherosclerotic lesions is largely derived from MCs and that activated MCs may contribute to atherogenesis by enhancing LDL retention.
Collapse
Affiliation(s)
- Katariina Maaninka
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Su Duy Nguyen
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Mikko I Mäyränpää
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland; Pathology, University of Helsinki and Helsinki University Hospital, Finland
| | - Riia Plihtari
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Kristiina Rajamäki
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland; Clinicum Department, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Perttu J Lindsberg
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland; Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Finland
| | - Petri T Kovanen
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Katariina Öörni
- Wihuri Research Institute, Biomedicum 1, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
13
|
Helin-Toiviainen M, Rönkkö S, Kaarniranta K, Puustjärvi T, Rekonen P, Ollikainen M, Uusitalo H. Oxidized low-density lipoprotein, lipid and calcium aggregates reveal oxidative stress and inflammation in the conjunctiva of glaucoma patients. Acta Ophthalmol 2017; 95:378-385. [PMID: 28139882 DOI: 10.1111/aos.13380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/08/2016] [Indexed: 12/26/2022]
Abstract
PURPOSE Conjunctival specimens from primary open-angle glaucoma (POAG), exfoliation glaucoma (ExG) patients and controls were histologically analysed for oxidized low-density lipoprotein (ox-LDL), lipid and calcium aggregates. Our goal was to use them as biomarkers of oxidative stress and inflammation and to evaluate their correlation with glaucoma and impact on surgical outcome. METHODS Conjunctival samples were obtained from POAG (n = 14) and ExG (n = 17) patients and from control subjects (n = 11) operated for macular hole, retinal detachment or strabismus. Immunohistochemistry was performed using the antibody against ox-LDL. Lipids and calcium were analysed by histochemical stainings with Nile red and Alizarin red S, respectively. RESULTS Immunoreaction for ox-LDL was significantly increased in POAG (p = 0.049) and the number of lipid aggregates was significantly higher in ExG (p = 0.009) when compared to control. When POAG and ExG patients were grouped according to the outcome of deep sclerectomy (DS) surgery, the number of lipid (p < 0.001) and calcium aggregates (p = 0.014) were significantly higher in the conjunctival stroma of patients whose surgery failed within a three-year follow-up period. CONCLUSIONS The lipid-mediated alterations suggested the presence of oxidative stress and inflammation in the conjunctiva of glaucoma patients. The present data further support the role of oxidative stress and inflammation in the wound healing process leading to excessive scarring and failure in DS surgery.
Collapse
Affiliation(s)
- Minna Helin-Toiviainen
- Department of Ophthalmology; Institute of Clinical Medicine; University of Eastern Finland; Kuopio Finland
| | - Seppo Rönkkö
- Faculty of Health Sciences; School of Pharmacy; Pharmaceutical Technology; University of Eastern Finland; Kuopio Finland
| | - Kai Kaarniranta
- Department of Ophthalmology; Institute of Clinical Medicine; University of Eastern Finland; Kuopio Finland
- Department of Ophthalmology; Kuopio University Hospital; Kuopio Finland
| | - Tuomo Puustjärvi
- Department of Ophthalmology; Kuopio University Hospital; Kuopio Finland
| | - Petri Rekonen
- Department of Ophthalmology; Institute of Clinical Medicine; University of Eastern Finland; Kuopio Finland
- Department of Ophthalmology; Kuopio University Hospital; Kuopio Finland
| | - Minna Ollikainen
- Department of Ophthalmology; Kuopio University Hospital; Kuopio Finland
| | - Hannu Uusitalo
- Department of Ophthalmology; University of Tampere; Tampere Finland
- Tampere University Hospital Eye Center; Medical School; University of Tampere; Tampere Finland
| |
Collapse
|
14
|
Sun CQ, Zhong CY, Sun WW, Xiao H, Zhu P, Lin YZ, Zhang CL, Gao H, Song ZY. Elevated Type II Secretory Phospholipase A2 Increases the Risk of Early Atherosclerosis in Patients with Newly Diagnosed Metabolic Syndrome. Sci Rep 2016; 6:34929. [PMID: 27941821 PMCID: PMC5150250 DOI: 10.1038/srep34929] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/20/2016] [Indexed: 12/24/2022] Open
Abstract
A critical association between type II secretory phospholipase A2 (sPLA2-IIa) and established atherosclerotic cardiovascular disease has been demonstrated. However, the contribution of sPLA2-IIa to early atherosclerosis remains unknown. This study investigated the association between early-stage atherosclerosis and sPLA2-IIa in metabolic syndrome (MetS) patients. One hundred and thirty-six MetS patients and 120 age- and gender-matched subjects without MetS were included. Serum sPLA2-IIa protein levels and activity were measured using commercial kits. Circulating endothelial activation molecules (vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), E-selectin, and P-selectin), and carotid intima-media thickness (cIMT), were measured as parameters of vascular endothelial dysfunction and early atherosclerosis. MetS patients exhibited significantly higher sPLA2-IIa protein and activity levels than the controls. Both correlated positively with fasting blood glucose and waist circumference in MetS patients. Additionally, MetS patients exhibited strikingly higher levels of endothelial activation molecules and increased cIMT than controls. These levels correlated positively with serum sPLA2-IIa protein levels and activity. Moreover, multivariate analysis showed that high sPLA2-IIa protein and activity levels were independent risk factors of early atherosclerosis in MetS patients. This study demonstrates an independent association between early-stage atherosclerosis and increased levels of sPLA2-IIa, implying that increased sPLA2-IIa may predict early-stage atherosclerosis in MetS patients.
Collapse
Affiliation(s)
- Chang-Qing Sun
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China.,Department of Geriatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Chun-Yan Zhong
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Wei-Wei Sun
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Hua Xiao
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Ping Zhu
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Yi-Zhang Lin
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Chen-Liang Zhang
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Hao Gao
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Zhi-Yuan Song
- Department of Cardiology, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
15
|
Hong CY, Han CT, Chao L. Nonspecific Binding Domains in Lipid Membranes Induced by Phospholipase A2. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:6991-6999. [PMID: 27218880 DOI: 10.1021/acs.langmuir.5b03915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Phospholipase A2 (PLA2) is a peripheral membrane protein that can hydrolyze phospholipids to produce lysolipids and fatty acids. It has been found to play crucial roles in various cellular processes and is thought as a potential candidate for triggering drug release from liposomes for medical treatment. Here, we directly observed that PLA2 hydrolysis reaction can induce the formation of PLA2-binding domains at lipid bilayer interface and found that the formation was significantly influenced by the fluidity of the lipid bilayer. We prepared supported lipid bilayers (SLBs) with various molar ratios of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) to adjust the reactivity and fluidity of the lipid bilayers. A significant amount of the PLA2-induced domains was observed in mixtures of DPPC and DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine) but not in either pure DPPC or pure DOPC bilayer, which might be the reason that previous studies rarely observed these domains in lipid bilayer systems. The fluorescently labeled PLA2 experiment showed that newly formed domains acted as binding templates for PLA2. The AFM result showed that the induced domain has stepwise plateau structure, suggesting that PLA2 hydrolysis products may align as bilayers and accumulate layer by layer on the support, and the hydrophobic acyl chains at the side of the layer structure may be exposed to the outside aqueous environment. The introduced hydrophobic region could have hydrophobic interactions with proteins and therefore can attract the binding of not only PLA2 but also other types of proteins such as proteoglycans and streptavidin. The results suggest that the formation of PLA2-induced domains may convert part of a zwitterionic nonsticky lipid membrane to a site where biomolecules can nonspecifically bind.
Collapse
Affiliation(s)
- Chia Yee Hong
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| | - Chung-Ta Han
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| | - Ling Chao
- Department of Chemical Engineering, National Taiwan University , Taipei, Taiwan 10617
| |
Collapse
|
16
|
Toba H, de Castro Brás LE, Baicu CF, Zile MR, Lindsey ML, Bradshaw AD. Increased ADAMTS1 mediates SPARC-dependent collagen deposition in the aging myocardium. Am J Physiol Endocrinol Metab 2016; 310:E1027-35. [PMID: 27143554 PMCID: PMC4935141 DOI: 10.1152/ajpendo.00040.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/29/2016] [Indexed: 01/08/2023]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a collagen-binding matricellular protein highly expressed during fibrosis. Fibrosis is a prominent component of cardiac aging that reduces myocardial elasticity. Previously, we reported that SPARC deletion attenuated myocardial stiffness and collagen deposition in aged mice. To investigate the mechanisms by which SPARC promotes age-related cardiac fibrosis, we evaluated six groups of mice (n = 5-6/group): young (3-5 mo old), middle-aged (10-12 mo old), and old (18-29 mo old) C57BL/6 wild type (WT) and SPARC-null (Null) mice. Collagen content, determined by picrosirius red staining, increased in an age-dependent manner in WT but not in Null mice. A disintegrin and metalloproteinase with thrombospondin-like motifs 1 (ADAMTS1) increased in middle-aged and old WT compared with young, whereas in Null mice only old animals showed increased ADAMTS1 expression. Versican, a substrate of ADAMTS1, decreased with age only in WT. To assess the mechanisms of SPARC-induced collagen deposition, we stimulated cardiac fibroblasts with SPARC. SPARC treatment increased secretion of collagen I and ADAMTS1 (both the 110-kDa latent and 87-kDa active forms) into the conditioned media as well as the cellular expression of transforming growth factor-β1-induced protein (Tgfbi) and phosphorylated Smad2. An ADAMTS1 blocking antibody suppressed the SPARC-induced collagen I secretion, indicating that SPARC promoted collagen production directly through ADAMTS1 interaction. In conclusion, ADAMTS1 is an important mediator of SPARC-regulated cardiac aging.
Collapse
Affiliation(s)
- Hiroe Toba
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; Department of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan;
| | - Lisandra E de Castro Brás
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; Department of Physiology, East Carolina University, Greenville, North Carolina
| | - Catalin F Baicu
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Michael R Zile
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, South Carolina; and
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; G. V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi
| | - Amy D Bradshaw
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, South Carolina; and
| |
Collapse
|
17
|
Thermal stability of human plasma electronegative low-density lipoprotein: A paradoxical behavior of low-density lipoprotein aggregation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1015-1024. [PMID: 27233433 DOI: 10.1016/j.bbalip.2016.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/08/2016] [Accepted: 05/21/2016] [Indexed: 12/20/2022]
Abstract
Low-density lipoprotein (LDL) aggregation is central in triggering atherogenesis. A minor fraction of electronegative plasma LDL, termed LDL(-), plays a special role in atherogenesis. To better understand this role, we analyzed the kinetics of aggregation, fusion and disintegration of human LDL and its fractions, LDL(+) and LDL(-). Thermal denaturation of LDL was monitored by spectroscopy and electron microscopy. Initially, LDL(-) aggregated and fused faster than LDL(+), but later the order reversed. Most LDL(+) disintegrated and precipitated upon prolonged heating. In contrast, LDL(-) partially retained lipoprotein morphology and formed soluble aggregates. Biochemical analysis of all fractions showed no significant degradation of major lipids, mild phospholipid oxidation, and an increase in non-esterified fatty acid (NEFA) upon thermal denaturation. The main baseline difference between LDL subfractions was higher content of NEFA in LDL(-). Since NEFA promote lipoprotein fusion, increased NEFA content can explain rapid initial aggregation and fusion of LDL(-) but not its resistance to extensive disintegration. Partial hydrolysis of apoB upon heating was similar in LDL subfractions, suggesting that minor proteins importantly modulate LDL disintegration. Unlike LDL(+), LDL(-) contains small amounts of apoA-I and apoJ. Addition of exogenous apoA-I to LDL(+) hampered lipoprotein aggregation, fusion and precipitation, while depletion of endogenous apoJ had an opposite effect. Therefore, the initial rapid aggregation of LDL(-) is apparently counterbalanced by the stabilizing effects of minor proteins such as apoA-I and apoJ. These results help identify key determinants for LDL aggregation, fusion and coalescence into lipid droplets in vivo.
Collapse
|
18
|
Zheng J, Zhou H, Zhao Y, Lun Q, Liu B, Tu P. Triterpenoid-enriched extract of Ilex kudingcha inhibits aggregated LDL-induced lipid deposition in macrophages by downregulating low density lipoprotein receptor-related protein 1 (LRP1). J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
19
|
Pop D, Dădârlat A, Zdrenghea D. Novel cardiovascular risk markers in women with ischaemic heart disease. Cardiovasc J Afr 2015; 25:137-41. [PMID: 25000444 PMCID: PMC4120125 DOI: 10.5830/cvja-2014-014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/14/2014] [Indexed: 11/06/2022] Open
Abstract
Abstract The incidence of coronary heart disease in premenopausal women is lower than in men because of their hormonal protection. Angina pectoris occurs in women about 10 years later than in men. However, mortality from ischaemic heart disease remains higher in women than in men. Current studies are focusing on novel cardiovascular risk biomarkers because it seems that traditional cardiovascular risk factors and their assessment scores underestimate the risk in females. Increased plasma levels of these newly established biomarkers of risk have been found to worsen endothelial dysfunction and inflammation, both of which play a key role in the pathogenesis of microvascular angina, which is very common in women. These novel cardiovascular risk markers can be classified into three categories: inflammatory markers, markers of haemostasis, and other biomarkers.
Collapse
Affiliation(s)
- Dana Pop
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania.
| | - Alexandra Dădârlat
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania
| | - D Zdrenghea
- University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Romania
| |
Collapse
|
20
|
Lu M, Gursky O. Aggregation and fusion of low-density lipoproteins in vivo and in vitro. Biomol Concepts 2015; 4:501-18. [PMID: 25197325 DOI: 10.1515/bmc-2013-0016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Low-density lipoproteins (LDLs, also known as 'bad cholesterol') are the major carriers of circulating cholesterol and the main causative risk factor of atherosclerosis. Plasma LDLs are 20- to 25-nm nanoparticles containing a core of cholesterol esters surrounded by a phospholipid monolayer and a single copy of apolipoprotein B (550 kDa). An early sign of atherosclerosis is the accumulation of LDL-derived lipid droplets in the arterial wall. According to the widely accepted 'response-to-retention hypothesis', LDL binding to the extracellular matrix proteoglycans in the arterial intima induces hydrolytic and oxidative modifications that promote LDL aggregation and fusion. This enhances LDL uptake by the arterial macrophages and triggers a cascade of pathogenic responses that culminate in the development of atherosclerotic lesions. Hence, LDL aggregation, fusion, and lipid droplet formation are important early steps in atherogenesis. In vitro, a variety of enzymatic and nonenzymatic modifications of LDL can induce these reactions and thereby provide useful models for their detailed analysis. Here, we summarize current knowledge of the in vivo and in vitro modifications of LDLs leading to their aggregation, fusion, and lipid droplet formation; outline the techniques used to study these reactions; and propose a molecular mechanism that underlies these pro-atherogenic processes. Such knowledge is essential in identifying endogenous and exogenous factors that can promote or prevent LDL aggregation and fusion in vivo and to help establish new potential therapeutic targets to decelerate or even block these pathogenic reactions.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Physiology and Biophysics, Boston University School of Medicine, W321, 700 Albany Street, Boston, MA 02118, USA.
| | | |
Collapse
|
21
|
Guijas C, Rodríguez JP, Rubio JM, Balboa MA, Balsinde J. Phospholipase A2 regulation of lipid droplet formation. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1841:1661-71. [PMID: 25450448 DOI: 10.1016/j.bbalip.2014.10.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/02/2014] [Accepted: 10/14/2014] [Indexed: 02/07/2023]
Abstract
The classical regard of lipid droplets as mere static energy-storage organelles has evolved dramatically. Nowadays these organelles are known to participate in key processes of cell homeostasis, and their abnormal regulation is linked to several disorders including metabolic diseases (diabetes, obesity, atherosclerosis or hepatic steatosis), inflammatory responses in leukocytes, cancer development and neurodegenerative diseases. Hence, the importance of unraveling the cell mechanisms controlling lipid droplet biosynthesis, homeostasis and degradation seems evident Phospholipase A2s, a family of enzymes whose common feature is to hydrolyze the fatty acid present at the sn-2 position of phospholipids, play pivotal roles in cell signaling and inflammation. These enzymes have recently emerged as key regulators of lipid droplet homeostasis, regulating their formation at different levels. This review summarizes recent results on the roles that various phospholipase A2 forms play in the regulation of lipid droplet biogenesis under different conditions. These roles expand the already wide range of functions that these enzymes play in cell physiology and pathophysiology.
Collapse
|
22
|
Sahmani M, Darabi M, Darabi M, Dabaghi T, Alizadeh SA, Najafipour R. The 763C>G Polymorphism of The Secretory PLA2IIa Gene Is Associated with Endometriosis in Iranian Women. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 8:437-44. [PMID: 25780526 PMCID: PMC4355930 DOI: 10.22074/ijfs.2015.4184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 01/28/2014] [Indexed: 11/15/2022]
Abstract
Background Endometriosis is a chronic gynecological disease resulting from complex
interactions between genetic, hormonal, environmental and oxidative stress and intrinsic
inflammatory components. The aim of this study was to investigate the potential association of the 763C>G polymorphism in the secretory phospholipase A2 group IIa gene
(PLA2G2A) with the risk of endometriosis in Iranian women. Materials and Methods Ninety seven patients with endometriosis along with 107 women who were negative for endometriosis after laparoscopy and laparatomy, and served as
the control group, were enrolled for this cross-sectional study. Samples were genotyped
using the polymerase chain reaction-restriction fragment length polymorphism method. Results Multivariate analysis was used to examine the association between the risk of endometriosis and the 763C>G polymorphism of PLA2G2A. Genotype distributions of PLA2G2A were significantly different between patients and the controls (p<0.001, OR=0.22, 95%
CI=0.21-0.39). Correlation analysis showed that there was a significant association between
the normal homozygous genotype and susceptibility to endometriosis (p<0.001). Conclusion The present study suggests that the 763C>G polymorphism of PLA2G2A plays
an important role as an independent factor in the risk of endometriosis in Iranian women.
Collapse
Affiliation(s)
- Mehdi Sahmani
- Department of Clinical Biochemistry and Medical Genetics, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Darabi
- Department of Clinical Biochemistry and Medical Genetics, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Safar Ali Alizadeh
- Department of Clinical Biochemistry and Medical Genetics, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Reza Najafipour
- Department of Clinical Biochemistry and Medical Genetics, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
23
|
Sun QA, Runge MS, Madamanchi NR. Oxidative stress, NADPH oxidases, and arteries. Hamostaseologie 2015; 36:77-88. [PMID: 25649240 DOI: 10.5482/hamo-14-11-0076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis and its major complications - myocardial infarction and stroke - remain major causes of death and disability in the United States and world-wide. Indeed, with dramatic increases in obesity and diabetes mellitus, the prevalence and public health impact of cardiovascular diseases (CVD) will likely remain high. Major advances have been made in development of new therapies to reduce the incidence of atherosclerosis and CVD, in particular for treatment of hypercholesterolemia and hypertension. Oxidative stress is the common mechanistic link for many CVD risk factors. However, only recently have the tools existed to study the interface between oxidative stress and CVD in animal models. The most important source of reactive oxygen species (and hence oxidative stress) in vascular cells are the multiple forms of enzymes nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase). Recently published and emerging studies now clearly establish that: 1) NADPH oxidases are of critical importance in atherosclerosis and hypertension in animal models; 2) given the tissue-specific expression of key components of NADPH oxidase, it may be possible to target vascular oxidative stress for prevention of CVD.
Collapse
Affiliation(s)
| | - Marschall S Runge
- Marschall S. Runge, MD PhD, Department of Medicine, 125 MacNider Hall, University of North Carolina, Chapel Hill, NC 27599-7005, USA, E-mail:
| | | |
Collapse
|
24
|
Shojaei Nik MH, Darabi M, Ziaee A, Hajmanoochehri F. Serum Phospholipase A2-IIA, hs-CRP, and Lipids in Women With Subclinical Hypothyroidism. Int J Endocrinol Metab 2014; 12:e16967. [PMID: 25237323 PMCID: PMC4166036 DOI: 10.5812/ijem.16967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 05/10/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Subclinical hypothyroidism (SCH) is a metabolic disorder characterized by elevated TSH level but normal T4 level. Some previous studies suggest that SCH is associated with inflammation. OBJECTIVES The present study aimed to compare lipid serum levels in SCH patients and normal participants, also explore possible association between SCH and the two inflammatory markers hs-CRP and PLA2-IIA. PATIENTS AND METHODS This study was performed on 77 women aged 20-45 (39 with SCH and 38 in the control group). TSH and T4 levels were measured by electrochemiluminescenceassay. Lipid profiles were analyzed using enzymatic-colorimetric methods. Hs-CRP and PLA2-IIA were determined using the ELISA method. IBM SPSS 19.0 was used for statistical analysis. RESULTS Serum levels of TG, cholesterol, and LDL were higher in the SCH group than the control group. However, there was no significant difference between the two groups for HDL level. Likewise, no difference was observed for the serum level of hs-CRP. PLA2-IIA mean value was higher in the SCH group. CONCLUSIONS SCH is associated with increased level of PLA2-IIA, which is independent of BMI. The stronger association of SCH with PLA2-IIA than with hs-CRP indicates that PLA2-IIA is an inducer of inflammation while hs-CRP is not.
Collapse
Affiliation(s)
- Mohammad Hossein Shojaei Nik
- Department of Biochemistry and Clinical Laboratories, International Branch (Aras), Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Masoud Darabi
- Liver and Gastrointestinal Disease Research Center, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, IR Iran
| | - Amir Ziaee
- Metabolic Diseases Research Center, Qazvin University of Medical Sciences, Qazvin, IR Iran
| | - Fatemeh Hajmanoochehri
- Metabolic Diseases Research Center, Qazvin University of Medical Sciences, Qazvin, IR Iran
- Corresponding author: Fatemeh Hajmanoochehri, Department of Pathology, Qazvin University of Medical Sciences, Booali-Sina St. Booali-Sina Hospital, Qazvin Metabolic Diseases Research Center, Qazvin, IR Iran. Tel: +98-2813360084, Fax: +98-2813326033, E-mail:
| |
Collapse
|
25
|
Lehti S, Käkelä R, Hörkkö S, Kummu O, Helske-Suihko S, Kupari M, Werkkala K, Kovanen PT, Öörni K. Modified lipoprotein-derived lipid particles accumulate in human stenotic aortic valves. PLoS One 2013; 8:e65810. [PMID: 23762432 PMCID: PMC3676354 DOI: 10.1371/journal.pone.0065810] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/29/2013] [Indexed: 01/22/2023] Open
Abstract
In aortic stenosis plasma lipoprotein-derived lipids accumulate in aortic valves. Here, we first compared the lipid compositions of stenotic aortic valves and atherosclerotic plaque cores. Both pathological tissues were found to be enriched in cholesteryl linoleate, a marker of extracellularly accumulated lipoproteins. In addition, a large proportion of the phospholipids were found to contain arachidonic acid, the common precursor of a number of proinflammatory lipid mediators. Next, we isolated and characterized extracellular lipid particles from human stenotic and non-stenotic control valves, and compared them to plasma lipoproteins from the same subjects. The extracellular valvular lipid particles were isolated from 15 stenotic and 14 non-stenotic aortic valves. Significantly more apoB-100-containing lipid particles were found in the stenotic than in the non-stenotic valves. The majority of the lipid particles isolated from the non-stenotic valves had sizes (23±6.2 nm in diameter) similar to those of plasma low density lipoprotein (LDL) (22±1.5 nm), while the lipid particles from stenotic valves were not of uniform size, their sizes ranging from 18 to more than 500 nm. The lipid particles showed signs of oxidative modifications, and when compared to isolated plasma LDL particles, the lipid particles isolated from the stenotic valves had a higher sphingomyelin/phosphatidylcholine –ratio, and also higher contents of lysophosphatidylcholine and unesterified cholesterol. The findings of the present study reveal, for the first time, that in stenotic human aortic valves, infiltrated plasma lipoproteins have undergone oxidative and lipolytic modifications, and become fused and aggregated. The generated large lipid particles may contribute to the pathogenesis of human aortic stenosis.
Collapse
Affiliation(s)
- Satu Lehti
- Wihuri Research Institute, Helsinki, Finland
| | - Reijo Käkelä
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Sohvi Hörkkö
- Institute of Diagnostics, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland
- Clinical Research Center, Oulu University Hospital, Oulu, Finland
- NordLab Oulu, Oulu University Hospital, Oulu, Finland
| | - Outi Kummu
- Institute of Diagnostics, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland
- Clinical Research Center, Oulu University Hospital, Oulu, Finland
| | - Satu Helske-Suihko
- Wihuri Research Institute, Helsinki, Finland
- Division of Cardiology, Helsinki University Central Hospital, Helsinki, Finland
| | - Markku Kupari
- Division of Cardiology, Helsinki University Central Hospital, Helsinki, Finland
| | - Kalervo Werkkala
- Division of Cardiothoracic Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | | | | |
Collapse
|
26
|
Gao X, Gong H, Men P, Zhou L, Ye D. Design, Synthesis, and Biological Evaluation of Novel Dual Inhibitors of Secretory Phospholipase A2 and Sphingomyelin Synthase. CHINESE J CHEM 2013. [DOI: 10.1002/cjoc.201300079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Lu M, Gantz DL, Herscovitz H, Gursky O. Kinetic analysis of thermal stability of human low density lipoproteins: a model for LDL fusion in atherogenesis. J Lipid Res 2012; 53:2175-2185. [PMID: 22855737 DOI: 10.1194/jlr.m029629] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fusion of modified LDL in the arterial wall promotes atherogenesis. Earlier we showed that thermal denaturation mimics LDL remodeling and fusion, and revealed kinetic origin of LDL stability. Here we report the first quantitative analysis of LDL thermal stability. Turbidity data show sigmoidal kinetics of LDL heat denaturation, which is unique among lipoproteins, suggesting that fusion is preceded by other structural changes. High activation energy of denaturation, E(a) = 100 ± 8 kcal/mol, indicates disruption of extensive packing interactions in LDL. Size-exclusion chromatography, nondenaturing gel electrophoresis, and negative-stain electron microscopy suggest that LDL dimerization is an early step in thermally induced fusion. Monoclonal antibody binding suggests possible involvement of apoB N-terminal domain in early stages of LDL fusion. LDL fusion accelerates at pH < 7, which may contribute to LDL retention in acidic atherosclerotic lesions. Fusion also accelerates upon increasing LDL concentration in near-physiologic range, which likely contributes to atherogenesis. Thermal stability of LDL decreases with increasing particle size, indicating that the pro-atherogenic properties of small dense LDL do not result from their enhanced fusion. Our work provides the first kinetic approach to measuring LDL stability and suggests that lipid-lowering therapies that reduce LDL concentration but increase the particle size may have opposite effects on LDL fusion.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118.
| | - Donald L Gantz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118
| | - Haya Herscovitz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118
| | - Olga Gursky
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118.
| |
Collapse
|
28
|
|
29
|
Leger AJ, Mosquea LM, Li L, Chuang W, Pacheco J, Taylor K, Luo Z, Piepenhagen P, Ziegler R, Moreland R, Urabe A, Jiang C, Cheng SH, Yew NS. Adeno-associated virus-mediated expression of acid sphingomyelinase decreases atherosclerotic lesion formation in apolipoprotein E−/− mice. J Gene Med 2011; 13:324-32. [DOI: 10.1002/jgm.1575] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
| | | | - Lingyun Li
- Genzyme Corporation; Framingham; MA; USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Azar M, Valentin E, Badaoui G, Kassab R, Sarkis A, Azar RR. Comparison of the effects of combination atorvastatin (40 mg) + ezetimibe (10 mg) versus atorvastatin (40 mg) alone on secretory phospholipase A2 activity in patients with stable coronary artery disease or coronary artery disease equivalent. Am J Cardiol 2011; 107:1571-4. [PMID: 21439529 DOI: 10.1016/j.amjcard.2011.01.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/20/2011] [Accepted: 01/20/2011] [Indexed: 10/18/2022]
Abstract
Secretory phospholipase A2 (sPLA2) is an enzyme that plays an important role in the pathogenesis of atherosclerosis and of adverse cardiovascular events. It is currently the target of emerging therapeutic agents. Our study was designed to investigate the effect of aggressive lowering of low-density lipoprotein (LDL) cholesterol with ezetimibe and atorvastatin on sPLA2 activity. We randomized 100 patients with stable coronary artery disease (CAD) or CAD equivalent (diabetes, stroke, or peripheral vascular disease) to receive ezetimibe 10 mg/day in association with atorvastatin 40 mg/day (combination therapy group) versus atorvastatin 40 mg/day and placebo (monotherapy group). Patients on statin therapy before inclusion were allowed to enter the study as long as the potency of the statin was lower than atorvastatin 40 mg/day. Lipid profile, high-sensitivity C-reactive protein (hs-CRP), and sPLA activity were measured at baseline and after 8 weeks of therapy. The decrease in LDL cholesterol was more significant in the combination therapy group, but the decrease in hs-CRP was similar. sPLA2 activity significantly decreased in the ezetimibe/atorvastatin group from 29 U/ml (interquartile range 23 to 35) to 26 U/ml (23 to 29, p = 0.001) but remained similar in the placebo/atorvastatin group (23 U/ml, 19 to 32, vs 22 U/ml, 19 to 28, p = NS). In a multivariate stepwise linear regression model, change in sPLA2 correlated with change in hs-CRP (p <0.001), baseline LDL cholesterol level (p = 0.001), body mass index (p = 0.003), diabetes mellitus (p = 0.04) and combination therapy with ezetimibe/atorvastatin (p = 0.05). In conclusion, this study demonstrates that coadministration of ezetimibe and atorvastatin decreases sPLA2 activity.
Collapse
|
31
|
Guha M, Gursky O. Effects of oxidation on structural stability and remodeling of human very low density lipoprotein. Biochemistry 2011; 49:9584-93. [PMID: 20919745 DOI: 10.1021/bi101391z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Very low density lipoproteins (VLDL) are triglyceride-rich precursors of low-density lipoproteins (LDL) and a risk factor for atherosclerosis. The effects of oxidation on VLDL metabolism may be pro- or antiatherogenic. To understand the underlying biophysical basis, we determined the effects of copper (that preferentially oxidizes lipids) and hypochlorite (that preferentially oxidizes proteins) on the heat-induced VLDL remodeling. This remodeling involves VLDL fusion, rupture, and fission of apoE-containing high-density lipoprotein- (HDL-) like particles; HDL with similar size, density, and protein composition are formed upon VLDL remodeling by lipoprotein lipase, a key enzyme in triglyceride metabolism. Circular dichroism, turbidity, and electron microscopy show that mild oxidation promotes VLDL fusion and rupture, while advanced oxidation hampers these reactions. VLDL destabilization upon moderate oxidation results, in part, from the exchangeable apolipoprotein modifications, including proteolysis and limited cross-linking. VLDL stabilization against fusion and rupture upon advanced oxidation probably results from massive protein cross-linking on the particle surface. Electron microscopy and gel electrophoresis reveal that oxidation promotes fission of apoE-containing HDL-size particles; hydrolysis of apolar core lipids probably contributes to this effect. Copper and hypochlorite have similar effects on VLDL remodeling, suggesting that these effects may be produced by other oxidants. In summary, moderate oxidation that encompasses in vivo conditions destabilizes VLDL and promotes fission of HDL-size particles. Consequently, mild oxidation may be synergistic with lipoprotein lipase reaction and, hence, may help to accelerate VLDL metabolism.
Collapse
|
32
|
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: 1.8] [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
|
33
|
Jayaraman S, Gantz DL, Gursky O. Effects of phospholipase A(2) and its products on structural stability of human LDL: relevance to formation of LDL-derived lipid droplets. J Lipid Res 2011; 52:549-57. [PMID: 21220788 DOI: 10.1194/jlr.m012567] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hydrolysis and oxidation of LDL stimulate LDL entrapment in the arterial wall and promote inflammation and atherosclerosis via various mechanisms including lipoprotein fusion and lipid droplet formation. To determine the effects of FFA on these transitions, we hydrolyzed LDL by phospholipase A(2) (PLA(2)), removed FFA by albumin, and analyzed structural stability of the modified lipoproteins. Earlier, we showed that heating induces LDL remodeling, rupture, and coalescence into lipid droplets resembling those found in atherosclerotic lesions. Here, we report how FFA affect these transitions. Circular dichroism showed that mild LDL lipolysis induces partial β-sheet unfolding in apolipoprotein B. Electron microscopy, turbidity, and differential scanning calorimetry showed that mild lipolysis promotes LDL coalescence into lipid droplets. FFA removal by albumin restores LDL stability but not the protein conformation. Consequently, FFA enhance LDL coalescence into lipid droplets. Similar effects of FFA were observed in minimally oxidized LDL, in LDL enriched with exogenous FFA, and in HDL and VLDL. Our results imply that FFA promote lipoprotein coalescence into lipid droplets and explain why LDL oxidation enhances such coalescence in vivo but hampers it in vitro. Such lipid droplet formation potentially contributes to the pro-atherogenic effects of FFA.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | |
Collapse
|
34
|
Rosenson RS. After FRANCIS: next steps in the clinical evaluation of varespladib methyl. Future Cardiol 2011; 7:11-8. [DOI: 10.2217/fca.10.118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Secretory phospholipase A2 (sPLA2) represents a family of isoenzymes that participate in lipoprotein and inflammatory pathways, mediate atherosclerosis and enhance myocardial ischemic injury. The Fewer Recurrent Acute Coronary Events with Near-term Cardiovascular Inflammatory Suppression (FRANCIS) trial (NCT00743925) was a Phase II trial designed to examine the effects of varespladib methyl, a small-molecule inhibitor of sPLA2, on plasma biomarkers in patients with acute coronary syndrome (ACS) who were treated with atorvastatin 80 mg and standard-of-care daily. Varespladib methyl significantly reduced low-density lipoprotein cholesterol and inflammatory biomarkers in ACS subjects treated with standard-of-care and atorvastatin 80 mg daily. There was a nonsignificant reduction in major adverse cardiovascular events at study completion; however, positive trends remained for unstable angina and myocardial infarction. In order to achieve the widespread use of varespladib methyl in ACS patients, completion of a prospective, randomized placebo-controlled trial in ACS patients and stable coronary artery disease patients with increased sPLA2 activity will be required.
Collapse
Affiliation(s)
- Robert S Rosenson
- Mount Sinai School of Medicine, Box 1030, One Gustave L Levy Place, New York, NY 10029, USA
| |
Collapse
|
35
|
Costales P, Aledo R, Vérnia S, Das A, Shah V, Casado M, Badimon L, Llorente-Cortés V. Selective role of sterol regulatory element binding protein isoforms in aggregated LDL-induced vascular low density lipoprotein receptor-related protein-1 expression. Atherosclerosis 2010; 213:458-68. [DOI: 10.1016/j.atherosclerosis.2010.09.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 09/15/2010] [Accepted: 09/29/2010] [Indexed: 11/24/2022]
|
36
|
Rosenson RS, Elliott M, Stasiv Y, Hislop C. Randomized trial of an inhibitor of secretory phospholipase A2 on atherogenic lipoprotein subclasses in statin-treated patients with coronary heart disease. Eur Heart J 2010; 32:999-1005. [PMID: 21081550 DOI: 10.1093/eurheartj/ehq374] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIMS To investigate the effects of secretory phospholipase A2 (sPLA(2)) inhibition on plasma lipoproteins. Secretory phospholipase A2 isoenzymes promote atherosclerosis by mechanisms that include lipoprotein modification, retention, and oxidation. METHODS AND RESULTS Phospholipase Levels And Serological Markers of Atherosclerosis II (PLASMA II) is a Phase II, randomized, double-blind, placebo-controlled parallel-arm study of two once-daily doses of the novel sPLA(2) inhibitor, 1-H-indole-3-glyoxamide or varespladib methyl (Anthera Pharmaceuticals, Hayward, CA, USA). One hundred and thirty-five stable coronary heart disease patients were treated with either varespladib methyl 250 mg once daily, varespladib methyl 500 mg once daily, or placebo for 8 weeks. Varespladib methyl treatment resulted in statistically significant dose-dependent reductions that were different from placebo in sPLA(2) concentration, low-density lipoprotein (LDL) cholesterol, and non-high-density lipoprotein (HDL) cholesterol. When compared with placebo, varespladib methyl 500 mg once daily reduced LDL cholesterol by 15% (P < 0.001), non-HDL cholesterol by 15% (P < 0.001), total very LDL (VLDL) particle concentration by 14% (P = 0.022), and small VLDL particle concentration by 24% (P = 0.030). Relative to baseline, varespladib methyl 500 mg once daily reduced total LDL particle concentration (7%, P = 0.002) and small LDL particle concentration (11%, P = 0.014). CONCLUSION Reductions in atherogenic lipoproteins suggest that varespladib methyl 500 mg once daily may be an effective anti-atherosclerotic agent. Trial registered at ClinicalTrials.gov, identifier: NCT00525954.
Collapse
Affiliation(s)
- Robert S Rosenson
- Mount Sinai Heart-Box 1030, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|
37
|
Rosenson RS, Hislop C, Elliott M, Stasiv Y, Goulder M, Waters D. Effects of varespladib methyl on biomarkers and major cardiovascular events in acute coronary syndrome patients. J Am Coll Cardiol 2010; 56:1079-88. [PMID: 20863951 DOI: 10.1016/j.jacc.2010.06.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Revised: 05/19/2010] [Accepted: 06/01/2010] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate the effects of varespladib on cardiovascular biomarkers in acute coronary syndrome patients. BACKGROUND Secretory phospholipase A(2) (sPLA(2)) represents a family of proatherogenic enzymes that hydrolyze lipoprotein phospholipids, increasing their affinity for intimal proteoglycans; contribute to cholesterol loading of macrophages by nonscavenger receptor mediated pathways; and activate inflammatory pathways. In prospective studies, high sPLA(2)-IIA levels predicted major adverse cardiovascular events in acute coronary syndrome (ACS) and stable coronary heart disease patients. METHODS This randomized, double-blind, prospective controlled clinical trial (phase 2B) was designed to investigate the effects of sPLA(2) inhibition with varespladib 500 mg daily versus placebo as adjunctive therapy to atorvastatin 80 mg daily on biomarkers (low-density lipoprotein cholesterol [LDL-C], high-sensitivity C-reactive protein [hsCRP], and sPLA(2)-IIA levels), major adverse cardiovascular events (unstable angina, myocardial infarction, death), and safety. In all, 625 ACS subjects were randomized within 96 h of the index event and treated for a minimum of 6 months. RESULTS After 8 weeks (primary efficacy end point), varespladib/atorvastatin reduced mean LDL-C levels from baseline by 49.6% compared with 43.4% with placebo/atorvastatin (p = 0.002). Respective 8-week median reductions in sPLA(2)-IIA levels were 82.4% and 15.6% (p < 0.0001), and hsCRP levels were lowered by 75.0% and 71.0% (p = 0.097). At 24 weeks, respective reductions with varespladib and placebo were as follows: LDL-C 43.5% versus 37.6% (p < 0.05), hsCRP 79.8% versus 77.0% (p = 0.02), and sPLA(2)-IIA 78.5% versus 6.4% (p < 0.0001). Major adverse cardiovascular events were not different from placebo 6 months post-randomization (7.3% varespladib vs. 7.7% placebo). No treatment differences in elevated liver function studies on >1 occasion were observed. CONCLUSIONS Varespladib therapy effectively reduced LDL-C and inflammatory biomarkers in ACS patients treated with conventional therapy including atorvastatin 80 mg daily. There were no treatment differences in clinical cardiovascular events. (FRANCIS [Fewer Recurrent Acute Coronary Events With Near-Term Cardiovascular Inflammation Suppression]-ACS Trial: A Study of the Safety and Efficacy of A 002 in Subjects With Acute Coronary Syndromes; NCT00743925).
Collapse
|
38
|
Garces F, López F, Niño C, Fernandez A, Chacin L, Hurt-Camejo E, Camejo G, Apitz-Castro R. High plasma phospholipase A2 activity, inflammation markers, and LDL alterations in obesity with or without type 2 diabetes. Obesity (Silver Spring) 2010; 18:2023-9. [PMID: 20134414 DOI: 10.1038/oby.2010.9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plasma phospholipases A(2) (PLA(2)) hydrolyze phospholipids of circulating lipoproteins or deposited in arteries producing bioactive lipids believed to contribute to the atherosclerotic inflammatory response. PLA(2)(s) are elevated in obesity and type 2 diabetes (T2D) but it is not clear which of these conditions is the cause since they frequently coexist. This study attempts to evaluate if high plasma PLA(2)(s) activities and markers of their effects in lipoproteins are associated with obesity or T2D diabetes, or with both. Total PLA(2) and Ca(2+)-dependent and -independent activities, lipids, lipoproteins, apoAI, and apoB apolipoproteins and affinity of apoB-lipoproteins for arterial proteoglycans were measured, as well as Inflammation markers. These parameters were evaluated in plasma samples of four groups: (i) apparently healthy controls with normal BMI (nBMI), (ii) obese subjects with no T2D, (iii) patients with T2D but with nBMI, and (iv) obese patients with T2D. PLA(2) activities were measured in the presence and absence of Ca(2+) and in the presence of specific inhibitors. Obese subjects, with or without T2D, had high activities of total PLA(2) and of Ca(2+)-dependent and Ca(2+)-independent enzymes. The activities were correlated with inflammation markers in obese subjects with and without diabetes and with alterations of low-density lipoproteins (LDLs) that increased their affinity for arterial proteoglycans. Ca(2+)-dependent secretory (sPLA(2)) enzymes were the main responsible of the obesity-associated high activity. We speculate that augmented PLA(2)(s) activity that increases affinity of circulating LDL for arterial intima proteoglycans could be another atherogenic component of obesity.
Collapse
Affiliation(s)
- Fatima Garces
- Laboratorio de Investigaciones Básicas y Aplicadas, Escuela de Bioanálisis, Facultad de Medicina, Universidad Central de Venezuela, Caracas, Venezuela
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Varespladib (A-002), a secretory phospholipase A2 inhibitor, reduces atherosclerosis and aneurysm formation in ApoE-/- mice. J Cardiovasc Pharmacol 2010; 53:60-5. [PMID: 19129734 DOI: 10.1097/fjc.0b013e318195bfbc] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The family of secretory phospholipase A2 (sPLA2) enzymes has been associated with inflammatory diseases and tissue injury including atherosclerosis. A-001 is a novel inhibitor of sPLA2 enzymes discovered by structure-based drug design, and A-002 is the orally bioavailable prodrug currently in clinical development. A-001 inhibited human and mouse sPLA2 group IIA, V, and X enzymes with IC50 values in the low nM range. A-002 (1 mg/kg) led to high serum levels of A-001 and inhibited PLA2 activity in transgenic mice overexpressing human sPLA2 group IIA in C57BL/6J background. In addition, the effects of A-002 on atherosclerosis in 2 ApoE mouse models were evaluated using en face analysis. (1) In a high-fat diet model, A-002 (30 and 90 mg/kg twice a day for 16 weeks) reduced aortic atherosclerosis by 50% (P < 0.05). Plasma total cholesterol was decreased (P < 0.05) by 1 month and remained lowered throughout the study. (2) In an accelerated atherosclerosis model, with angiotensin II-induced aortic lesions and aneurysms, A-002 (30 mg/kg twice a day) reduced aortic atherosclerosis by approximately 40% (P < 0.05) and attenuated aneurysm formation (P = 0.0096). Thus, A-002 was effective at significantly decreasing total cholesterol, atherogenesis, and aneurysm formation in these 2 ApoE mouse models.
Collapse
|
40
|
Karakas M, Koenig W. Phospholipase A2 as a therapeutic target for atherosclerosis. ACTA ACUST UNITED AC 2010. [DOI: 10.2217/clp.09.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Ishikawa Y, Kimura-Matsumoto M, Murakami M, Murakami M, Yamamoto K, Akasaka Y, Uzuki M, Yuri Y, Inomata N, Yokoo T, Ishii T. Distribution of smooth muscle cells and macrophages expressing scavenger receptor BI/II in atherosclerosis. J Atheroscler Thromb 2009; 16:829-39. [PMID: 20032583 DOI: 10.5551/jat.1941] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Scavenger receptors type I and II (SRBI/II) have dual roles in both atherogenic and antiatherogenic functions through interactions with lipoproteins and their expression in macrophages; how-ever, the distribution and density of SRBI/II-positive macrophages and smooth muscle cells (SMCs) as well as their association with lipid metabolism-related proteins in atherosclerotic intima of the human aorta remain unclear. METHODS Autopsied aortic tissues were double-immunostained with SRBI/BII and smooth muscle actin or macrophage-specific antibodies. The density of SRBI/BII-positive SMCs and macrophages in intimal lesion was measured. They were also immunostained with antibodies against four apolipoproteins, four phospholipase A2s, and CETP. RESULTS SRBI/II was expressed in both macrophages and SMCs distributed in various intimal lesions. The density of SRBI/II-positive SMCs in intimal lesions significantly decreased with the advance of atherosclerosis, whereas the density of SRBI/II-positive macrophages significantly increased with atherosclerotic development. In addition, functional proteins, such as apolipoproteins, secretory phospholipase A2s, and CETP, were distributed in the intimal stroma around SRBI/II-positive cells in all lesion types. CONCLUSION The results indicated that SMCs are involved in lipid metabolism via SRBI/II expression mainly in the early stages of atherosclerosis evolution, and that SRBI/II-positive macrophages are mainly involved in advanced stages.
Collapse
Affiliation(s)
- Yukio Ishikawa
- Department of Pathology, Toho University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Abstract
The initial event in atherogenesis is the increased transcytosis of low density lipoprotein, and its subsequent deposition, retention and modification in the subendothelium. It is followed by the infiltration of activated inflammatory cells from the coronary circulation into the arterial wall. There they secrete reactive oxygen species (ROS) and produce oxidized lipoproteins capable of inducing endothelial cell apoptosis, and thereby plaque erosion. Activated T lymphocytes, macrophages and mast cells, accumulate in the eroded plaque where they secrete a variety of proteases capable of inducing degradation of extracellular proteins, thereby rendering the plaques more prone to rupture. This review summarizes the recent advancements in the understanding of the roles of ROS and oxidized lipoproteins in the activation of inflammatory cells and inducing signalling pathways related to cell death and apoptosis. In addition, it presents evidence that this vicious circle between oxidative stress and inflammation does not only occur in the diseased arterial wall, but also in adipose tissues. There, oxidative stress and inflammation impair adipocyte maturation resulting in defective insulin action and adipocytokine signalling. The latter is associated with increased infiltration of inflammatory cells, loss of anti-oxidant protection and cell death in the arterial wall.
Collapse
Affiliation(s)
- Maarten Hulsmans
- Atherosclerosis and Metabolism Unit, Department of Cardiovascular Diseases, Katholieke Universiteit Leuven, Belgium
| | | |
Collapse
|
44
|
Hulsmans M, Holvoet P. The vicious circle between oxidative stress and inflammation in atherosclerosis. FASEB J 2009; 25:2515-27. [PMID: 19968738 DOI: 10.1096/fj.11-181149] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The initial event in atherogenesis is the increased transcytosis of low density lipoprotein, and its subsequent deposition, retention and modification in the subendothelium. It is followed by the infiltration of activated inflammatory cells from the coronary circulation into the arterial wall. There they secrete reactive oxygen species (ROS) and produce oxidized lipoproteins capable of inducing endothelial cell apoptosis, and thereby plaque erosion. Activated T lymphocytes, macrophages and mast cells, accumulate in the eroded plaque where they secrete a variety of proteases capable of inducing degradation of extracellular proteins, thereby rendering the plaques more prone to rupture. This review summarizes the recent advancements in the understanding of the roles of ROS and oxidized lipoproteins in the activation of inflammatory cells and inducing signalling pathways related to cell death and apoptosis. In addition, it presents evidence that this vicious circle between oxidative stress and inflammation does not only occur in the diseased arterial wall, but also in adipose tissues. There, oxidative stress and inflammation impair adipocyte maturation resulting in defective insulin action and adipocytokine signalling. The latter is associated with increased infiltration of inflammatory cells, loss of anti-oxidant protection and cell death in the arterial wall.
Collapse
Affiliation(s)
- Maarten Hulsmans
- Atherosclerosis and Metabolism Unit, Department of Cardiovascular Diseases, Katholieke Universiteit Leuven, Belgium
| | | |
Collapse
|
45
|
van der Giet M, Tölle M, Pratico D, Lufft V, Schuchardt M, Hörl MP, Zidek W, Tietge UJF. Increased type IIA secretory phospholipase A(2) expression contributes to oxidative stress in end-stage renal disease. J Mol Med (Berl) 2009; 88:75-83. [PMID: 19798476 DOI: 10.1007/s00109-009-0543-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 09/06/2009] [Accepted: 09/07/2009] [Indexed: 12/26/2022]
Abstract
End-stage renal disease (ESRD) patients exhibit increased in vivo oxidative stress conceivably contributing to cardiovascular mortality. The type IIA secretory phospholipase A(2) (sPLA(2)) has proatherogenic activity. We explored the hypothesis that sPLA(2) contributes to oxidative stress generation and endothelial dysfunction in ESRD patients and transgenic (tg) mice. Patients with ESRD had increased in vivo oxidative stress as assessed by plasma isoprostane levels (p < 0.001). Active sPLA(2) in plasma was substantially increased compared with healthy controls (1,156 +/- 65 versus 184 +/- 5 ng/dL, p < 0.001) and correlated with plasma isoprostanes (r = 0.61, p < 0.001). Correspondingly, human sPLA(2) tg mice display increased generation of reactive oxygen species within aortic vascular smooth muscle cells, leading to severe endothelial dysfunction (maximal vasodilation in response to 10 micromol/L acetylcholine, sPLA(2) 36 +/- 8%, controls 80 +/- 2% of phenylephrine-induced vasoconstriction). Increased vascular oxidative stress in sPLA(2) tg mice is dependent on the induction of vascular cyclooxygenase (COX)2 expression. Conversely, ESRD patients show increased formation of COX2-derived prostaglandins (p < 0.05) correlated with plasma sPLA(2) (r = 0.71, p < 0.05). Our data indicate that increased expression of sPLA(2) might represent a novel causative risk factor contributing to the increased cardiovascular disease morbidity and mortality in ESRD.
Collapse
Affiliation(s)
- Markus van der Giet
- Medizinische Klinik IV-Nephrology, Charite-Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Oörni K, Kovanen PT. Lipoprotein modification by secretory phospholipase A(2) enzymes contributes to the initiation and progression of atherosclerosis. Curr Opin Lipidol 2009; 20:421-7. [PMID: 19593123 DOI: 10.1097/mol.0b013e32832fa14d] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Secretory phospholipase A2s (sPLA2s) are considered to be important enzymes in the initiation and progression of atherosclerosis. In this review, we discuss the various mechanisms by which the direct action of the sPLA2s on LDL particles in the arterial intima may contribute to atherogenesis. RECENT FINDINGS A wealth of evidence, both in vitro and in vivo, supports a role for the sPLA2s in atherogenesis. Very recently, systemic inhibition of sPLA2s was found to reduce measures of arterial inflammation. The mechanisms behind this inhibition, however, are largely unknown. Here, we discuss the consequences of sPLA2 action on LDL in the arterial intima and address the recent findings regarding the effects of the lipolytic products of sPLA2, lysophosphatidylcholine, and fatty acids on intimal cells. LDL modified by sPLA2 can accumulate in the arterial intima both extracellularly and intracellularly. Importantly, the lipolytic products promote atherosclerosis by monocyte/macrophage recruitment, by enhancing the production of proretentive molecules by vascular smooth muscle cells, and by inducing cell death. SUMMARY Recent findings on sPLA2s support the idea that the enzymes contribute to human atherogenesis not only as initiating agents but also in maintaining plaque inflammation.
Collapse
|
47
|
Schmitz G, Ruebsaamen K. Metabolism and atherogenic disease association of lysophosphatidylcholine. Atherosclerosis 2009; 208:10-8. [PMID: 19570538 DOI: 10.1016/j.atherosclerosis.2009.05.029] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 04/27/2009] [Accepted: 05/25/2009] [Indexed: 10/20/2022]
Abstract
Lysophosphatidylcholine (LPC) is a major plasma lipid that has been recognized as an important cell signalling molecule produced under physiological conditions by the action of phospholipase A(2) on phosphatidylcholine. LPC transports glycerophospholipid components such as fatty acids, phosphatidylglycerol and choline between tissues. LPC is a ligand for specific G protein-coupled signalling receptors and activates several second messengers. LPC is also a major phospholipid component of oxidized low-density lipoproteins (Ox-LDL) and is implicated as a critical factor in the atherogenic activity of Ox-LDL. Hence, LPC plays an important role in atherosclerosis and acute and chronic inflammation. In this review we focus in some detail on LPC function, biochemical pathways, sources and signal-transduction system. Moreover, we outline the detection of LPC by mass spectrometry which is currently the best method for accurate and simultaneous analysis of each individual LPC species and reveal the pathophysiological implication of LPC which makes it an interesting target for biomarker and drug development regarding atherosclerosis and cardiovascular disorders.
Collapse
Affiliation(s)
- Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|
48
|
Shetty R, Pibarot P, Audet A, Janvier R, Dagenais F, Perron J, Couture C, Voisine P, Després JP, Mathieu P. Lipid-mediated inflammation and degeneration of bioprosthetic heart valves. Eur J Clin Invest 2009; 39:471-80. [PMID: 19490057 DOI: 10.1111/j.1365-2362.2009.02132.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND The durability of bioprosthetic valves is limited by structural valve degeneration (SVD) leading to bioprostheses (BPs) stenosis or regurgitation. We hypothesized that a lipid-mediated inflammatory mechanism is involved in the SVD of BPs. MATERIAL AND METHODS Eighteen Freestyle stentless BP valves were explanted for SVD at a mean time of 5.9 +/- 3 years after implantation and were analysed by immunohistochemistry and transmission electron microscopy (TEM). RESULTS The mean age of the patients was 65 +/- 8 years and there were 11 male and seven female patients. Two of the 18 BPs had macroscopic calcification, whereas the other valves had minimal or no macroscopic calcification. Tears at the commissures leading to regurgitation was present in 16 BPs. Immunohistochemistry showed the presence of oxidized low-density lipoprotein (ox-LDL) and glycosaminoglycans in the fibrosa layer of 13 BPs. Areas with ox-LDL were infiltrated by macrophages (CD68(+)) co-expressing the scavenger receptor CD36 and metalloproteinase-9 (MMP-9). Zymogram showed the active form of MMP-9 within explanted BPs. EM studies revealed the presence of lipid-laden cells featuring foam cells and fragmented collagen. Nonimplanted control BPs obtained from the manufacturer (n = 4) had no evidence of lipid accumulation, inflammatory cell infiltration or expression of MMP9 within the leaflets. CONCLUSIONS These results support the concept that lipid-mediated inflammatory mechanisms may contribute to the SVD of BPs. These findings suggest that modification of atherosclerotic risk factors with the use of behavioural or pharmacological interventions could help to reduce the incidence of SVD.
Collapse
Affiliation(s)
- R Shetty
- Laval University, Quebec, QC, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lima LM, das Graças Carvalho M, da Fonseca Neto CP, Garcia JCF, Sousa MO. Secretory phospholipase A2 in patients with coronary artery disease. J Thromb Thrombolysis 2009; 29:276-81. [DOI: 10.1007/s11239-009-0345-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Accepted: 04/30/2009] [Indexed: 10/20/2022]
|
50
|
Rosenson RS, Hislop C, McConnell D, Elliott M, Stasiv Y, Wang N, Waters DD. Effects of 1-H-indole-3-glyoxamide (A-002) on concentration of secretory phospholipase A2 (PLASMA study): a phase II double-blind, randomised, placebo-controlled trial. Lancet 2009; 373:649-58. [PMID: 19231633 DOI: 10.1016/s0140-6736(09)60403-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Secretory phospholipase A(2) (sPLA(2)) enzymes, produced and secreted in human blood vessels and hepatocytes, contribute to the development of atherosclerosis through mechanisms that are both dependent and independent of lipoprotein. We examined the effects of an sPLA(2) inhibitor on enzyme concentration and on plasma lipoproteins and inflammatory biomarkers in patients with coronary heart disease. METHODS Patients aged 18 years and older with stable coronary heart disease from the USA and Ukraine were eligible for enrolment in this phase II, randomised, double-blind, placebo-controlled, parallel-arm, dose-response study. 393 patients were randomly assigned by computer-generated sequence to receive either placebo (n=79) or one of four doses of an sPLA(2) inhibitor, A-002 (1-H-indole-3-glyoxamide; 50 mg [n=79], 100 mg [n=80], 250 mg [n=78], or 500 mg [n=77] twice daily), for 8 weeks. The primary endpoint was the change in sPLA(2) group IIA (sPLA(2)-IIA) concentration or activity from baseline to week 8. Analysis was by modified intention to treat (ITT). The ITT population consisted of all patients who received one dose of study treatment; data for patients who dropped out before the end of the study were carried forward from last observation. This trial is registered with ClinicalTrials.gov, number NCT00455546. FINDINGS All randomised patients received at least one dose and were included in the ITT population. Data for 45 patients were carried forward from last observation (36 in the A-002 group and nine in the placebo group); the main reason for dropout before completion was because of adverse events. 348 patients reached the primary endpoint (A-002 n=278, placebo n=70). Mean sPLA(2)-IIA concentration fell by 86.7%, from 157 pmol/L to 21 [corrected] pmol/L, in the overall active treatment group, and by 4.8%, from 157 pmol/L to 143 [corrected] pmol/L, in the placebo group (p<0.0001 treatment vs placebo). The reductions in sPLA(2)-IIA concentration in the A-002 groups were dose dependent (ranging from 69.2% in the 50 mg group to 95.8% in the 500 mg group) and differed significantly from placebo (p<0.0001 for all doses). In the 500 mg A-002 treatment group, there was one serious adverse event (exacerbation of underlying chronic obstructive pulmonary disease), but the proportion of patients reporting treatment-emergent adverse events did not differ from placebo. The main side-effects of the drug included headache (n=20), nausea (n=17), and diarrhoea (n=12). INTERPRETATION The reductions in sPLA(2)-IIA concentration suggest that A-002 might be an effective anti-atherosclerotic agent.
Collapse
Affiliation(s)
- Robert S Rosenson
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | |
Collapse
|