1
|
Ahmed IU, Myerscough MR. HDL and plaque regression in a multiphase model of early atherosclerosis. Math Biosci 2024; 373:109208. [PMID: 38759951 DOI: 10.1016/j.mbs.2024.109208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Atherosclerosis is a chronic disease of the arteries characterised by the accumulation of lipids and lipid-engorged cells in the artery wall. Early plaque growth is aggravated by the deposition of low density lipoproteins (LDL) in the wall and the subsequent immune response. High density lipoproteins (HDL) counterbalance the effects of LDL by accepting cholesterol from macrophages and removing it from the plaque. In this paper, we develop a free boundary multiphase model to investigate the effects of LDL and HDL on early plaque development. We examine how the rates of LDL and HDL deposition affect cholesterol accumulation in macrophages, and how this impacts cell death rates and emigration. We identify a region of LDL-HDL parameter space where plaque growth stabilises for low LDL and high HDL influxes, due to macrophage emigration and HDL clearance that counterbalances the influx of new cells and cholesterol. We explore how the efferocytic uptake of dead cells and the recruitment of new macrophages affect plaque development for a range of LDL and HDL influxes. Finally, we consider how changes in the LDL-HDL profile can change the course of plaque development. We show that changes towards lower LDL and higher HDL can slow plaque growth and even induce regression. We find that these changes have less effect on larger, more established plaques, and that temporary changes will only slow plaque growth in the short term.
Collapse
Affiliation(s)
- Ishraq U Ahmed
- School of Mathematics and Statistics, University of Sydney, Australia.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Australia
| |
Collapse
|
2
|
Machlovi SI, Neuner SM, Hemmer BM, Khan R, Liu Y, Huang M, Zhu JD, Castellano JM, Cai D, Marcora E, Goate AM. APOE4 confers transcriptomic and functional alterations to primary mouse microglia. Neurobiol Dis 2022; 164:105615. [PMID: 35031484 PMCID: PMC8934202 DOI: 10.1016/j.nbd.2022.105615] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 12/09/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Common genetic variants in more than forty loci modulate risk for Alzheimer's disease (AD). AD risk alleles are enriched within enhancers active in myeloid cells, suggesting that microglia, the brain-resident macrophages, may play a key role in the etiology of AD. A major genetic risk factor for AD is Apolipoprotein E (APOE) genotype, with the ε4/ε4 (E4) genotype increasing risk for AD by approximately 15 fold compared to the most common ε3/ε3 (E3) genotype. However, the impact of APOE genotype on microglial function has not been thoroughly investigated. To address this, we cultured primary microglia from mice in which both alleles of the mouse Apoe gene have been humanized to encode either human APOE ε3 or APOE ε4. Relative to E3 microglia, E4 microglia exhibit altered morphology, increased endolysosomal mass, increased cytokine/chemokine production, and increased lipid and lipid droplet accumulation at baseline. These changes were accompanied by decreased translation and increased phosphorylation of eIF2ɑ and eIF2ɑ-kinases that participate in the integrated stress response, suggesting that E4 genotype leads to elevated levels of cellular stress in microglia relative to E3 genotype. Using live-cell imaging and flow cytometry, we also show that E4 microglia exhibited increased phagocytic uptake of myelin and other substrates compared to E3 microglia. While transcriptomic profiling of myelin-challenged microglia revealed a largely overlapping response profile across genotypes, differential enrichment of genes in interferon signaling, extracellular matrix and translation-related pathways was identified in E4 versus E3 microglia both at baseline and following myelin challenge. Together, our results suggest E4 genotype confers several important functional alterations to microglia even prior to myelin challenge, providing insight into the molecular and cellular mechanisms by which APOE4 may increase risk for AD.
Collapse
Affiliation(s)
- Saima I Machlovi
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah M Neuner
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brittany M Hemmer
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Riana Khan
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min Huang
- James J Peters VA Medical Center, Research & Development, Bronx, NY, USA; Department of Neurology, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeffrey D Zhu
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph M Castellano
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Department of Neurology, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dongming Cai
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; James J Peters VA Medical Center, Research & Development, Bronx, NY, USA; Department of Neurology, New York, NY, USA; Alzheimer Disease Research Center, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, USA; Department of Genetics and Genomic Sciences, New York, NY, USA; Department of Neurology, New York, NY, USA; Alzheimer Disease Research Center, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Han YS, Chen JX, Li ZB, Chen J, Yi WJ, Huang H, Wei LL, Jiang TT, Li JC. Identification of potential lipid biomarkers for active pulmonary tuberculosis using ultra-high-performance liquid chromatography-tandem mass spectrometry. Exp Biol Med (Maywood) 2020; 246:387-399. [PMID: 33175608 DOI: 10.1177/1535370220968058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Early diagnosis of active pulmonary tuberculosis (TB) is the key to controlling the disease. Host lipids are nutrient sources for the metabolism of Mycobacterium tuberculosis. In this research work, we used ultra-high-performance liquid chromatography-tandem mass spectrometry to screen plasma lipids in TB patients, lung cancer patients, community-acquired pneumonia patients, and normal healthy controls. Principal component analysis, orthogonal partial least squares discriminant analysis, and K-means clustering algorithm analysis were used to identify lipids with differential abundance. A total of 22 differential lipids were filtered out among all subjects. The plasma phospholipid levels were decreased, while the cholesterol ester levels were increased in patients with TB. We speculate that the infection of M. tuberculosis may regulate the lipid metabolism of TB patients and may promote host-assisted bacterial degradation of phospholipids and accumulation of cholesterol esters. This may be related to the formation of lung cavities with caseous necrosis. The results of receiver operating characteristic curve analysis revealed four lipids such as phosphatidylcholine (PC, 12:0/22:2), PC (16:0/18:2), cholesteryl ester (20:3), and sphingomyelin (d18:0/18:1) as potential biomarkers for early diagnosis of TB. The diagnostic model was fitted by using logistic regression analysis and combining the above four lipids with a sensitivity of 92.9%, a specificity of 82.4%, and the area under the curve (AUC) value of 0.934 (95% CI 0.873 - 0.971). The machine learning method (10-fold cross-validation) demonstrated that the model had good accuracy (0.908 AUC, 85.3% sensitivity, and 85.9% specificity). The lipids identified in this study may serve as novel biomarkers in TB diagnosis. Our research may pave the foundation for understanding the pathogenesis of TB.
Collapse
Affiliation(s)
- Yu-Shuai Han
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jia-Xi Chen
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhi-Bin Li
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Chen
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wen-Jing Yi
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.,Central Laboratory, Yangjiang People's Hospital, Yangjiang 529500, China
| | - Huai Huang
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.,Central Laboratory, Yangjiang People's Hospital, Yangjiang 529500, China
| | - Li-Liang Wei
- Department of Pneumology, Shaoxing Municipal Hospital, Shaoxing 312000, China
| | - Ting-Ting Jiang
- Central Laboratory, Yangjiang People's Hospital, Yangjiang 529500, China
| | - Ji-Cheng Li
- Institute of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
4
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
5
|
Elyasi A, Voloshyna I, Ahmed S, Kasselman LJ, Behbodikhah J, De Leon J, Reiss AB. The role of interferon-γ in cardiovascular disease: an update. Inflamm Res 2020; 69:975-988. [PMID: 32699989 DOI: 10.1007/s00011-020-01382-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is the leading cause of death, globally, and its prevalence is only expected to rise due to the increasing incidence of co-morbidities such as obesity and diabetes. Medical treatment of CVD is directed primarily at slowing or reversing the underlying atherosclerotic process by managing circulating lipids with an emphasis on control of low-density lipoprotein (LDL) cholesterol. However, over the past several decades, there has been increasing recognition that chronic inflammation and immune system activation are important contributors to atherosclerosis. This shift in focus has led to the elucidation of the complex interplay between cholesterol and cellular secretion of cytokines involved in CVD pathogenesis. Of the vast array of cytokine promoting atherosclerosis, interferon (IFN)-γ is highly implicated and, therefore, of great interest. METHODS Literature review was performed to further understand the effect of IFN-γ on the development of atherosclerotic CVD. RESULTS IFN-γ, the sole member of the type II IFN family, is produced by T cells and macrophages, and has been found to induce production of other cytokines and to have multiple effects on all stages of atherogenesis. IFN-γ activates a variety of signaling pathways, most commonly the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, to induce oxidative stress, promote foam cell accumulation, stimulate smooth muscle cell proliferation and migration into the arterial intima, enhance platelet-derived growth factor expression, and destabilize plaque. These are just a few of the contributions of IFN-γ to the initiation and progression of atherosclerotic CVD. CONCLUSION Given the pivotal role of IFN-γ in the advancement of CVD, activation of its signaling pathways is being explored as a driver of atherosclerosis. Manipulation of this key cytokine may lead to novel therapeutic avenues for CVD prevention and treatment. A number of therapies are being explored with IFN-γ as the potential target.
Collapse
Affiliation(s)
- Ailin Elyasi
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Iryna Voloshyna
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Saba Ahmed
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Lora J Kasselman
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Jennifer Behbodikhah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Allison B Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| |
Collapse
|
6
|
Macrophage polarisation associated with atherosclerosis differentially affects their capacity to handle lipids. Atherosclerosis 2020; 305:10-18. [PMID: 32592946 DOI: 10.1016/j.atherosclerosis.2020.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 04/02/2020] [Accepted: 05/07/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Lipid-rich foam cell macrophages drive atherosclerosis via several mechanisms, including inflammation, lipid uptake, lipid deposition and plaque vulnerability. The atheroma environment shapes macrophage function and phenotype; anti-inflammatory macrophages improve plaque stability while pro-inflammatory macrophages promote rupture. Current evidence suggests a variety of macrophage phenotypes occur in atherosclerotic plaques with local lipids, cytokines, oxidised phospholipids and pathogenic stimuli altering their phenotype. In this study, we addressed differential functioning of macrophage phenotypes via a systematic analysis of in vitro polarised, human monocyte-derived macrophage phenotypes, focussing on molecular events that regulate foam-cell formation. METHODS We examined transcriptomes, protein levels and functionally determined lipid handling and foam cell formation capacity in macrophages polarised with IFNγ+LPS, IL-4, IL-10, oxPAPC and CXCL4. RESULTS RNA sequencing of differentially polarised macrophages revealed distinct gene expression changes, with enrichment in atherosclerosis and lipid-associated pathways. Analysis of lipid processing activity showed IL-4 and IL-10 macrophages have higher lipid uptake and foam cell formation activities, while inflammatory and oxPAPC macrophages displayed lower foam cell formation. Inflammatory macrophages showed low lipid uptake, while higher lipid uptake in oxPAPC macrophages was matched by increased lipid efflux capacity. CONCLUSIONS Atherosclerosis-associated macrophage polarisation dramatically affects lipid handling capacity underpinned by major transcriptomic changes and altered protein levels in lipid-handling gene expression. This leads to phenotype-specific differences in LDL uptake, cellular cholesterol levels and cholesterol efflux, informing how the plaque environment influences atherosclerosis progression by influencing macrophage phenotypes.
Collapse
|
7
|
Zhou QD, Chi X, Lee MS, Hsieh WY, Mkrtchyan JJ, Feng AC, He C, York AG, Bui VL, Kronenberger EB, Ferrari A, Xiao X, Daly AE, Tarling EJ, Damoiseaux R, Scumpia PO, Smale ST, Williams KJ, Tontonoz P, Bensinger SJ. Interferon-mediated reprogramming of membrane cholesterol to evade bacterial toxins. Nat Immunol 2020; 21:746-755. [PMID: 32514064 PMCID: PMC7778040 DOI: 10.1038/s41590-020-0695-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022]
Abstract
Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.
Collapse
Affiliation(s)
- Quan D Zhou
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Xun Chi
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Min Sub Lee
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wei Yuan Hsieh
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan J Mkrtchyan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - An-Chieh Feng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cuiwen He
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Autumn G York
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| | - Viet L Bui
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eliza B Kronenberger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Allison E Daly
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elizabeth J Tarling
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Philip O Scumpia
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kevin J Williams
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Steven J Bensinger
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Shin G, Jang K, Kim M, Lee JH, Yoo HJ. Inflammatory Markers and Plasma Fatty Acids in Predicting WBC Level Alterations in Association With Glucose-Related Markers: A Cross-Sectional Study. Front Immunol 2020; 11:629. [PMID: 32346379 PMCID: PMC7172801 DOI: 10.3389/fimmu.2020.00629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/19/2020] [Indexed: 12/26/2022] Open
Abstract
Aging leads to immune function changes which contribute to occurrence of chronic conditions. White blood cell (WBC) level is a marker widely known to reflect the immune function, thus, prediction of WBC level changes by using certain biomarkers is needed to prevent chronic conditions and to decrease the burdens of aging. In this respect, the present study aimed to explore the relationships between inflammatory markers and plasma fatty acid (FA) composition according to WBC levels for verifying potential predictors of WBC levels. Study subjects were divided into three groups according to their WBC count: moderate-low WBC (MLW), normal WBC, and moderate-high WBC (MHW). Inflammatory markers were measured, and plasma FA profiles were constructed via gas chromatography-mass spectrometry (GC-MS). In the MHW group, insulin, homeostatic model assessment of insulin resistance (HOMA-IR), γ-glutamyltransferase (GGT), high-sensitivity C-reactive protein (hs-CRP), and interferon (IFN)-γ showed significant increases compared to those in the other groups. In addition, the granulocyte-to-lymphocyte ratio (GLR) significantly increased according to the WBC levels, whereas the platelet-to-lymphocyte ratio (PLR) showed the opposite result. Total ω-3 polyunsaturated fatty acids (PUFAs) showed significant differences among the groups. Regarding ω-6 PUFAs, dihomo-γ-linolenic acid and docosatetraenoic acid levels were significantly increased in the MHW group compared to the other groups. Finally, multivariate linear regression analysis revealed that GGT, hs-CRP, IFN-γ, ω-3 PUFAs, and dihomo-γ-linolenic acid were independent factors for altering WBC levels. In conclusion, elevated WBC levels accompanied by an increased GLR and a decreased PLR were associated with the risk of type 2 diabetes based on increased insulin and HOMA-IR levels and decreased adiponectin levels. Additionally, GGT, hs-CRP, IFN-γ, ω-3 PUFAs, and dihomo-γ-linolenic acid levels emerged as independent biomarkers for predicting WBC level alterations. Therefore, this study showed that these inflammatory markers and plasma FAs not only affect WBC level alterations but also may play roles in the risk of type 2 diabetes as one of the chronic conditions by certain mechanisms, which should be further studied. Finally, checking these biomarkers along with WBC levels can be helpful to prevent the chronic conditions.
Collapse
Affiliation(s)
- Gurum Shin
- Department of Food and Nutrition, College of Human Ecology, National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomic, Yonsei University, Seoul, South Korea
| | - Kyunghye Jang
- Department of Food and Nutrition, College of Human Ecology, National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomic, Yonsei University, Seoul, South Korea
| | - Minjoo Kim
- Department of Food and Nutrition, College of Life Science and Nano Technology, Hannam University, Daejeon, South Korea
| | - Jong Ho Lee
- Department of Food and Nutrition, College of Human Ecology, National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomic, Yonsei University, Seoul, South Korea.,Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Hye Jin Yoo
- Department of Food and Nutrition, College of Human Ecology, National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomic, Yonsei University, Seoul, South Korea.,Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| |
Collapse
|
9
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
10
|
Rahmati-Ahmadabad S, Broom DR, Ghanbari-Niaki A, Shirvani H. Effects of exercise on reverse cholesterol transport: A systemized narrative review of animal studies. Life Sci 2019; 224:139-148. [PMID: 30922848 DOI: 10.1016/j.lfs.2019.03.058] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 12/20/2022]
Abstract
AIMS Reverse Cholesterol Transport (RCTr) is the mechanism by which excess cholesterol from peripheral tissues is transported to the liver for hepatobiliary excretion, thereby inhibiting foam cell formation and the development of atherosclerosis. Exercise affects RCTr, by influencing high-density lipoprotein cholesterol (HDL) through remodeling and by promoting hepatobiliary sterol excretion. The objectives of this systematized review of animal studies is to summarize the literature and provide an overview of the effects of chronic exercise (at least two weeks) on apolipoproteins (Apo A-I, Apo-E), Paraoxonase-1 (PON1), ATP-binding cassette transporters (ABCA1, ABCG1, ABCG4, ABCG5, ABCG8), scavenger receptor class B type I (SR-BI), cholesteryl ester transfer protein (CETP), low-density lipoprotein receptor (LDLr) and cholesterol 7 alpha-hydroxylase (CYP7A1) and Niemann-Pick C1-like 1 (NPC1L1). MATERIALS AND METHODS Three electronic databases (PubMed, Science Direct and Google Scholar) were searched for eligible studies conducted from the earliest available date to August 2018. KEY FINDINGS Most of studies investigate the effects of low to moderate intensity aerobic training on RCTr elements. The majority were on exercised rats undertaking moderate intensity aerobic training. SIGNIFICANCE This review highlights that moderate intensity and longer-term training has a greater effect on RCTr elements than low intensity training. There a few studies examining high intensity training which warrants further investigation.
Collapse
Affiliation(s)
| | - David Robert Broom
- Academy of Sport and Physical Activity, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, UK
| | - Abbass Ghanbari-Niaki
- Exercise Biochemistry Division, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Mazandaran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
CD36 in chronic kidney disease: novel insights and therapeutic opportunities. Nat Rev Nephrol 2017; 13:769-781. [DOI: 10.1038/nrneph.2017.126] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
12
|
Kumar M, Coburn J, Kaplan DL, Mandal BB. Immuno-Informed 3D Silk Biomaterials for Tailoring Biological Responses. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29310-29322. [PMID: 27726371 DOI: 10.1021/acsami.6b09937] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Macrophages, the key players in immunoregulation, are actively involved in tissue remodelling and vascularization. Recent advances in tissue engineering and regenerative medicine illustrate the importance of "immuno-informed" biomaterials to regulate the microenvironment of biomedical implants. In the current study, silk-based 3D hydrogels were utilized to regulate cytokine delivery for macrophage, a type of immune cell, differentiation and polarization. Three different hydrogel variants, silk-poly(ethylene glycol) (PEG) (SP), silk-horseradish peroxidase (HRP) (SH) and silk-sonicated (SS) hydrogels were studied. Hydrogels were loaded with the M1 and M2 polarizing cytokines interferon-γ (IFN-γ) and interleukin-4 (IL-4), respectively. Functional cytokine release and macrophage polarization studies were conducted using three cytokine exposure approaches: only cytokine encapsulation (macrophage in culture well), only macrophage encapsulation (cytokine in culture media) and cytokine with macrophage encapsulation. The extent of macrophage polarization by cytokine-eluting and macrophage-encapsulating hydrogels was investigated using gene expression analysis for C-C chemokine receptor 7 (CCR7), Interleukin-1 beta (IL-1β), cluster of differentiation 206 (CD206) and cluster of differentiation 209 (CD209). The released cytokines polarized macrophages from an M0 phenotype to an M1/M2 phenotype. Also, lineage committed M1/M2 macrophages could be "switched" to their M2/M1 counterparts (M1-to-M2 or M2-to-M1 transition) exhibiting their well-established plasticity. When macrophages were encapsulated in hydrogels, polarization could be induced to the lineage committed M1 or M2 phenotypes either in polarizing media or when coencapsulated with cytokines. Through this study, silk hydrogels demonstrated utility as a novel system for focal delivery of cytokines and macrophages as "immuno-informed" 3D silk-biomaterials.
Collapse
Affiliation(s)
- Manishekhar Kumar
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG) , Guwahati, 781039, India
| | - Jeannine Coburn
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts United States
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG) , Guwahati, 781039, India
| |
Collapse
|
13
|
Immune-inflammatory responses in atherosclerosis: Role of an adaptive immunity mainly driven by T and B cells. Immunobiology 2016; 221:1014-33. [PMID: 27262513 DOI: 10.1016/j.imbio.2016.05.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 01/22/2023]
Abstract
Adaptive immune response plays an important role in atherogenesis. In atherosclerosis, the proinflammatory immune response driven by Th1 is predominant but the anti-inflammatory response mediated mainly by regulatory T cells is also present. The role of Th2 and Th17 cells in atherogenesis is still debated. In the plaque, other T helper cells can be observed such as Th9 and Th22 but is little is known about their impact in atherosclerosis. Heterogeneity of CD4(+) T cell subsets presented in the plaque may suggest for plasticity of T cell that can switch the phenotype dependening on the local microenvironment and activating/blocking stimuli. Effector T cells are able to recognize self-antigens released by necrotic and apoptotic vascular cells and induce a humoral immune reaction. Tth cells resided in the germinal centers help B cells to switch the antibody class to the production of high-affinity antibodies. Humoral immunity is mediated by B cells that release antigen-specific antibodies. A variety of B cell subsets were found in human and murine atherosclerotic plaques. In mice, B1 cells could spontaneously produce atheroprotective natural IgM antibodies. Conventional B2 lymphocytes secrete either proatherogenic IgG, IgA, and IgE or atheroprotective IgG and IgM antibodies reactive with oxidation-specific epitopes on atherosclerosis-associated antigens. A small population of innate response activator (IRA) B cells, which is phenotypically intermediate between B1 and B2 cells, produces IgM but possesses proatherosclerotic properties. Finally, there is a minor subset of splenic regulatory B cells (Bregs) that protect against atherosclerotic inflammation through support of generation of Tregs and production of anti-inflammatory cytokines IL-10 and TGF-β and proapoptotic molecules.
Collapse
|
14
|
|
15
|
Reeves ARD, Spiller KL, Freytes DO, Vunjak-Novakovic G, Kaplan DL. Controlled release of cytokines using silk-biomaterials for macrophage polarization. Biomaterials 2015; 73:272-83. [PMID: 26421484 DOI: 10.1016/j.biomaterials.2015.09.027] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 12/17/2022]
Abstract
Polarization of macrophages into an inflammatory (M1) or anti-inflammatory (M2) phenotype is important for clearing pathogens and wound repair, however chronic activation of either type of macrophage has been implicated in several diseases. Methods to locally control the polarization of macrophages is of great interest for biomedical implants and tissue engineering. To that end, silk protein was used to form biopolymer films that release either IFN-γ or IL-4 to control the polarization of macrophages. Modulation of the solubility of the silk films through regulation of β-sheet (crystalline) content enabled a short-term release (4-8 h) of either cytokine, with smaller amounts released out to 24 h. Altering the solubility of the films was accomplished by varying the time that the films were exposed to water vapor. The released IFN-γ or IL-4 induced polarization of THP-1 derived macrophages into the M1 or M2 phenotypes, respectively. The silk biomaterials were able to release enough IFN-γ or IL-4 to repolarize the macrophage from M1 to M2 and vice versa, demonstrating the well-established plasticity of macrophages. High β-sheet content films that are not soluble and do not release the trapped cytokines were also able to polarize macrophages that adhered to the surface through degradation of the silk protein. Chemically conjugating IFN-γ to silk films through disulfide bonds allowed for longer-term release to 10 days. The release of covalently attached IFN-γ from the films was also able to polarize M1 macrophages in vitro. Thus, the strategy described here offers new approaches to utilizing biomaterials for directing the polarization of macrophages.
Collapse
Affiliation(s)
- Andrew R D Reeves
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| | - Kara L Spiller
- School of Biomedical Engineering, Drexel University, Philadelphia, PA, USA.
| | - Donald O Freytes
- New York Stem Cell Foundation Research Institute, New York, NY, USA.
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|
16
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 347] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
17
|
Boshuizen MCS, de Winther MPJ. Interferons as Essential Modulators of Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1579-88. [PMID: 25953648 DOI: 10.1161/atvbaha.115.305464] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Abstract
Interferons (IFNs) are key regulators of both innate and adaptive immune responses. The family of IFN cytokines can be divided into 3 main subtypes of which type I and type II IFNs are most well-defined. IFNs are known to be important mediators in atherosclerosis. Evidence from both in vitro and in vivo studies shows that the IFNs are generally proatherosclerotic. However, their role in atherosclerosis is complex, with distinct roles for these cytokines throughout different stages of the disease. In this review, we will discuss the current knowledge on the role of type I and type II IFNs in atherosclerosis development, specifically focusing on their role in endothelial activation, cell recruitment, foam cell formation, and regulation of apoptosis. Furthermore, we will discuss whether IFNs could be considered as new molecular targets for therapeutic intervention in atherosclerosis.
Collapse
Affiliation(s)
- Marieke C S Boshuizen
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Menno P J de Winther
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
18
|
Yu XH, Zhang J, Zheng XL, Yang YH, Tang CK. Interferon-γ in foam cell formation and progression of atherosclerosis. Clin Chim Acta 2015; 441:33-43. [DOI: 10.1016/j.cca.2014.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 11/28/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
|
19
|
Thirunavukkarasu S, de Silva K, Plain KM, J Whittington R. Role of host- and pathogen-associated lipids in directing the immune response in mycobacterial infections, with emphasis on Mycobacterium avium subsp. paratuberculosis. Crit Rev Microbiol 2014; 42:262-75. [PMID: 25163812 DOI: 10.3109/1040841x.2014.932327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mycobacteria have a complex cell wall with a high lipid content that confers unique advantages for bacterial survival in the hostile host environment, leading to long-term infection. There is a wealth of evidence suggesting the role cell wall-associated lipid antigens play at the host-pathogen interface by contributing to bacterial virulence. One pathway that pathogenic mycobacteria use to subvert host immune pathways to their advantage is host cholesterol/lipid homeostasis. This review focuses on the possible role of pathogen- and host-associated lipids in the survival and persistence of pathogenic mycobacteria with emphasis on Mycobacterium avium subsp. paratuberculosis. We draw upon literature in diverse areas of infectious and metabolic diseases and explain a mechanism by which mycobacterial-induced changes in the host cellular energy state could account for phenomena that are a hallmark of chronic mycobacterial diseases.
Collapse
Affiliation(s)
| | - Kumudika de Silva
- a Faculty of Veterinary Science , University of Sydney , Camden , Australia
| | - Karren M Plain
- a Faculty of Veterinary Science , University of Sydney , Camden , Australia
| | | |
Collapse
|
20
|
Stolzing A, Sethe S, Grune T. Chronically active: activation of microglial proteolysis in ageing and neurodegeneration. Redox Rep 2013; 10:207-13. [PMID: 16259788 DOI: 10.1179/135100005x70198] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
One of the microglial cell functions is the removal of modified extracellular proteins in the brain. The connection between protein oxidation, proteolysis, and microglial activation is the topic of this review. The effect of various activation agents on microglial cells with regard to changes in substrate uptake, proteolytic capacity and degradation efficiency of different types of oxidized protein materials is reviewed. It is shown that different activation stimuli initiate substrate-specific modulation for uptake and proteolysis, influencing an array of factors including receptor expression, lysosomal pH, and proteasome subunit composition. Age-related alterations in activation and proteolytic capacity in microglial cells are also discussed. In ageing, proteolytic effectiveness is diminished, while microglial cells are chronically activated and lose the oxidative burst ability, possibly supporting a 'vicious circle' of macrophage-induced neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Centre for Biomaterials and Tissue Engineering, Sheffield University, UK
| | | | | |
Collapse
|
21
|
Wouters K, Cudejko C, Gijbels MJJ, Fuentes L, Bantubungi K, Vanhoutte J, Dièvart R, Paquet C, Bouchaert E, Hannou SA, Gizard F, Tailleux A, de Winther MPJ, Staels B, Paumelle R. Bone marrow p16INK4a-deficiency does not modulate obesity, glucose homeostasis or atherosclerosis development. PLoS One 2012; 7:e32440. [PMID: 22403661 PMCID: PMC3293804 DOI: 10.1371/journal.pone.0032440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/31/2012] [Indexed: 12/31/2022] Open
Abstract
Objective A genomic region near the CDKN2A locus, encoding p16INK4a, has been associated to type 2 diabetes and atherosclerotic vascular disease, conditions in which inflammation plays an important role. Recently, we found that deficiency of p16INK4a results in decreased inflammatory signaling in murine macrophages and that p16INK4a influences the phenotype of human adipose tissue macrophages. Therefore, we investigated the influence of immune cell p16INK4a on glucose tolerance and atherosclerosis in mice. Methods and Results Bone marrow p16INK4a-deficiency in C57Bl6 mice did not influence high fat diet-induced obesity nor plasma glucose and lipid levels. Glucose tolerance tests showed no alterations in high fat diet-induced glucose intolerance. While bone marrow p16INK4a-deficiency did not affect the gene expression profile of adipose tissue, hepatic expression of the alternative markers Chi3l3, Mgl2 and IL10 was increased and the induction of pro-inflammatory Nos2 was restrained on the high fat diet. Bone marrow p16INK4a-deficiency in low density lipoprotein receptor-deficient mice did not affect western diet-induced atherosclerotic plaque size or morphology. In line, plasma lipid levels remained unaffected and p16INK4a-deficient macrophages displayed equal cholesterol uptake and efflux compared to wild type macrophages. Conclusion Bone marrow p16INK4a-deficiency does not affect plasma lipids, obesity, glucose tolerance or atherosclerosis in mice.
Collapse
Affiliation(s)
- Kristiaan Wouters
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Céline Cudejko
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Marion J. J. Gijbels
- Departments of Molecular Genetics and Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Lucia Fuentes
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Kadiombo Bantubungi
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Jonathan Vanhoutte
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Rebecca Dièvart
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Charlotte Paquet
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Emmanuel Bouchaert
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Sarah Anissa Hannou
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Florence Gizard
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Anne Tailleux
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Menno P. J. de Winther
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Bart Staels
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
- * E-mail:
| | - Réjane Paumelle
- Univ Lille Nord de France, Lille, France
- Inserm, U1011, Lille, France
- Université Droit et Santé de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
22
|
Allahverdian S, Pannu PS, Francis GA. Contribution of monocyte-derived macrophages and smooth muscle cells to arterial foam cell formation. Cardiovasc Res 2012; 95:165-72. [PMID: 22345306 DOI: 10.1093/cvr/cvs094] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Smooth muscle cells (SMCs) are the main cell type in intimal thickenings and some stages of human atherosclerosis. Like monocyte-derived macrophages, SMCs accumulate excess lipids and contribute to the total intimal foam cell population. In contrast, apolipoprotein (Apo)E-deficient and LDL receptor-deficient mice develop atherosclerotic lesions that are macrophage- as opposed to SMC-rich. The lesser contribution of SMCs to lesion development in these mouse models has distracted attention away from the importance of SMC cholesterol homeostasis in the artery wall. Intimal SMCs accumulate excess amounts of cholesteryl esters when compared with medial layer SMCs, possibly explained by reduced ATP-binding cassette transporter A1 expression and ApoA-I binding to intimal-type SMCs. The aim of this review is to compare the relative contribution of monocyte-derived macrophages and SMCs to human vs. mouse atherosclerosis, and describe what is known about lipid uptake and removal mechanisms contributing to arterial macrophage and SMC foam cell formation. An increased understanding of the contribution of these cell types to lesion development will help to delineate their relative importance in atherogenesis and as potential therapeutic targets.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, UBC James Hogg Research Centre, Providence Heart + Lung Institute at St Paul's Hospital, Room 166, Burrard Building, 1081 Burrard Street, Vancouver, BC, Canada V6Z 1Y6
| | | | | |
Collapse
|
23
|
Wolfs IMJ, Donners MMPC, de Winther MPJ. Differentiation factors and cytokines in the atherosclerotic plaque micro-environment as a trigger for macrophage polarisation. Thromb Haemost 2011; 106:763-71. [PMID: 21947328 DOI: 10.1160/th11-05-0320] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 08/23/2011] [Indexed: 12/13/2022]
Abstract
The phenotype of macrophages in atherosclerotic lesions can vary dramatically, from a large lipid laden foam cell to a small inflammatory cell. Classically, the concept of macrophage heterogeneity discriminates between two extremes called either pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. Polarisation of plaque macrophages is predominantly determined by the local micro-environment present in the atherosclerotic lesion and is rather more complex than typically described by the M1/M2 paradigm. In this review we will discuss the role of various polarising factors in regulating the phenotypical state of plaque macrophages. We will focus on two main levels of phenotype regulation, one determined by differentiation factors produced in the lesion and the other determined by T-cell-derived polarising cytokines. With foam cell formation being a key characteristic of macrophages during atherosclerosis initiation and progression, these polarisation factors will also be linked to lipid handling of macrophages.
Collapse
Affiliation(s)
- I M J Wolfs
- Dept. of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | | | | |
Collapse
|
24
|
McLaren JE, Michael DR, Ashlin TG, Ramji DP. Cytokines, macrophage lipid metabolism and foam cells: implications for cardiovascular disease therapy. Prog Lipid Res 2011; 50:331-47. [PMID: 21601592 DOI: 10.1016/j.plipres.2011.04.002] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 04/14/2011] [Accepted: 04/14/2011] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease is the biggest killer globally and the principal contributing factor to the pathology is atherosclerosis; a chronic, inflammatory disorder characterized by lipid and cholesterol accumulation and the development of fibrotic plaques within the walls of large and medium arteries. Macrophages are fundamental to the immune response directed to the site of inflammation and their normal, protective function is harnessed, detrimentally, in atherosclerosis. Macrophages contribute to plaque development by internalizing native and modified lipoproteins to convert them into cholesterol-rich foam cells. Foam cells not only help to bridge the innate and adaptive immune response to atherosclerosis but also accumulate to create fatty streaks, which help shape the architecture of advanced plaques. Foam cell formation involves the disruption of normal macrophage cholesterol metabolism, which is governed by a homeostatic mechanism that controls the uptake, intracellular metabolism, and efflux of cholesterol. It has emerged over the last 20 years that an array of cytokines, including interferon-γ, transforming growth factor-β1, interleukin-1β, and interleukin-10, are able to manipulate these processes. Foam cell targeting, anti-inflammatory therapies, such as agonists of nuclear receptors and statins, are known to regulate the actions of pro- and anti-atherogenic cytokines indirectly of their primary pharmacological function. A clear understanding of macrophage foam cell biology will hopefully enable novel foam cell targeting therapies to be developed for use in the clinical intervention of atherosclerosis.
Collapse
Affiliation(s)
- James E McLaren
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | | | | | | |
Collapse
|
25
|
Chiurchiù V, Izzi V, D'Aquilio F, Vismara D, Carotenuto F, Catanzaro G, Maccarrone M. Endomorphin-1 prevents lipid accumulation via CD36 down-regulation and modulates cytokines release from human lipid-laden macrophages. Peptides 2011; 32:80-5. [PMID: 20932867 DOI: 10.1016/j.peptides.2010.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 09/24/2010] [Accepted: 09/24/2010] [Indexed: 12/21/2022]
Abstract
CD36 is a scavenger receptor known to play a critical role in the development of atherosclerosis by mediating the uptake of oxidized low-density lipoproteins (oxLDL) by macrophages, thus leading to foam cell formation. It is now generally recognized that the immune system has a pivotal role in the pathogenesis of atherosclerosis, whose progression is determined by ongoing inflammatory reactions. Recently, several studies pointed out that opioid peptides exert anti-inflammatory activities. Therefore the aim of the present study was to evaluate a possible endomorphin-1 (EM-1) immunomodulatory activity on human foam cells. Our results showed that EM-1 reduced Nile Red-stained lipid droplets content, decreased the expression of CD36 receptor and modulated tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) release from lipid-laden macrophages. Furthermore, Naloxone, an opioid receptors antagonist, reverted the anti-atherogenic and anti-inflammatory observed effects of EM-1. These data demonstrated, for the first time, an unprecedented ability of EM-1 to act as a novel modulator for macrophage-to-foam cell transformation, and for inflammatory cytokines profile, suggesting possible novel endomorphin-based anti-atherosclerotic approaches for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Valerio Chiurchiù
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC)/IRCCS Fondazione S.Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
26
|
McLaren JE, Michael DR, Salter RC, Ashlin TG, Calder CJ, Miller AM, Liew FY, Ramji DP. IL-33 Reduces Macrophage Foam Cell Formation. THE JOURNAL OF IMMUNOLOGY 2010; 185:1222-9. [DOI: 10.4049/jimmunol.1000520] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
Barnholt KE, Kota RS, Aung HH, Rutledge JC. Adenosine blocks IFN-gamma-induced phosphorylation of STAT1 on serine 727 to reduce macrophage activation. THE JOURNAL OF IMMUNOLOGY 2009; 183:6767-77. [PMID: 19846878 DOI: 10.4049/jimmunol.0900331] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Macrophages are activated by IFN-gamma, a proinflammatory and proatherogenic cytokine that mediates its downstream effects primarily through STAT1. IFN-gamma signaling induces phosphorylation of two STAT1 residues: Tyr(701) (Y701), which facilitates dimerization, nuclear translocation, and DNA binding; and Ser(727) (S727), which enables maximal STAT1 transcription activity. Immunosuppressive molecules such as adenosine in the cellular microenvironment can reduce macrophage inflammatory and atherogenic functions through receptor-mediated signaling pathways. We hypothesized that adenosine achieves these protective effects by interrupting IFN-gamma signaling in activated macrophages. This investigation demonstrates that adding adenosine to IFN-gamma-stimulated murine RAW 264.7 and human THP-1 macrophages results in unique modulation of STAT1 serine and tyrosine phosphorylation events. We show that adenosine inhibits IFN-gamma-induced STAT1 S727 phosphorylation by >30% and phosphoserine-mediated transcriptional activity by 58% but has no effect on phosphorylation of Y701 or receptor-associated JAK tyrosine kinases. Inhibition of the adenosine A(3) receptor with a subtype-specific antagonist (MRS 1191 in RAW 264.7 cells and MRS 1220 in THP-1 cells) reverses this adenosine suppressive effect on STAT1 phosphoserine status by 25-50%. Further, RAW 264.7 A(3) receptor stimulation with Cl-IB-MECA reduces IFN-gamma-induced STAT1 transcriptional activity by 45% and STAT1-dependent gene expression by up to 80%. These data suggest that A(3) receptor signaling is key to adenosine-mediated STAT1 modulation and anti-inflammatory action in IFN-gamma-activated mouse and human macrophages. Because STAT1 plays a key role in IFN-gamma-induced inflammation and foam cell transformation, a better understanding of the mechanisms underlying STAT1 deactivation by adenosine may improve preventative and therapeutic approaches to vascular disease.
Collapse
Affiliation(s)
- Kimberly E Barnholt
- Department of Internal Medicine, Division of Endocrinology, Clinical Nutrition, and Vascular Medicine, University of California, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
28
|
Hao XR, Cao DL, Hu YW, Li XX, Liu XH, Xiao J, Liao DF, Xiang J, Tang CK. IFN-γ down-regulates ABCA1 expression by inhibiting LXRα in a JAK/STAT signaling pathway-dependent manner. Atherosclerosis 2009; 203:417-28. [DOI: 10.1016/j.atherosclerosis.2008.07.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 07/08/2008] [Accepted: 07/24/2008] [Indexed: 12/30/2022]
|
29
|
Rogers L, Burchat S, Gage J, Hasu M, Thabet M, Willcox L, Wilcox L, Ramsamy TA, Whitman SC. Deficiency of invariant V alpha 14 natural killer T cells decreases atherosclerosis in LDL receptor null mice. Cardiovasc Res 2008; 78:167-74. [PMID: 18192239 PMCID: PMC5439367 DOI: 10.1093/cvr/cvn005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aims CD1d-restricted natural killer T (NKT) cells function by regulating numerous immune responses during innate and adaptive immunity. Depletion of all populations of CD1d-dependent NKT cells has been shown by several groups to reduce atherosclerosis in two different mouse models of the disease. In this study, we determined if removal of a single (Vα14) NKT cell population protects mice from the disease. Methods and results Targeted deletion of the Jα18 gene results in selective depletion of CD1d-dependent Vα14 NKT cells in C57BL/6 mice without affecting the population of other NKT, NK, and conventional T cells. Therefore, to study the effect of Vα14 NKT cell depletion on the progression of atherosclerosis, we examined the extent of lesion formation using paired littermate LDL receptor null mice that were either +/+ or −/− for the Jα18 gene following the feeding of these mice a cholesterol- and fat-enriched diet for 8 weeks. At the end of the study, we found no difference in either serum total- or lipoprotein-cholesterol distributions between groups. However, quantification of atherosclerosis revealed that Vα14 NKT cell deficiency significantly decreased lesion size in the aortic root (20–28%) and arch (28–38%) in both genders of mice. By coupling the techniques of laser capture microdissection with quantitative real-time RT–PCR, we found that expression of the proatherogenic cytokine interferon (IFN)-γ was significantly reduced in lesions from Jα18−/− mice. Conclusion This study is the first to identify a specific subpopulation of NKT cells that promotes atherosclerosis via a mechanism appearing to involve IFN-γ expression.
Collapse
Affiliation(s)
- Leah Rogers
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Agrawal S, Febbraio M, Podrez E, Cathcart MK, Stark GR, Chisolm GM. Signal transducer and activator of transcription 1 is required for optimal foam cell formation and atherosclerotic lesion development. Circulation 2007; 115:2939-47. [PMID: 17533179 DOI: 10.1161/circulationaha.107.696922] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Signal transducer and activator of transcription 1 (Stat1) potently regulates gene expression after stimulation by certain cytokines involved in tumorigenesis and host defenses. The present study investigated a novel role for Stat1 in foam cell formation and atherosclerosis. METHODS AND RESULTS Inhibition of Stat1 activity by a Stat1-specific DNA "decoy" oligomer transfected into differentiated human THP-1 cells, and deficiency of stat1 in mouse macrophages significantly inhibited foam cell formation assessed by lipid staining and cholesteryl ester accumulation compared with control cells. The mechanism of Stat1 regulation of foam cell formation was uniquely dependent on the scavenger receptor CD36. Blunted Stat1 activity and stat1 deficiency significantly decreased expression of CD36 but not of scavenger receptor-A compared with controls, as assessed by immunoblotting and flow cytometry. Deficiency of CD36 but not scavenger receptor-A in mouse macrophages removed any dependency of foam cell formation on Stat1. In an intraperitoneal model of foam cell formation in which foam cells form in vivo independently of the model ligands used in vitro, stat1 deficiency significantly inhibited foam cell formation and CD36 expression. Transplantation of bone marrow from apolipoprotein e-/- x stat1-/- mice into lethally irradiated, atherosclerosis-susceptible apolipoprotein e-/- recipients significantly reduced both en face aortic lesion coverage and aortic root lesions compared with recipients of bone marrow from genetically matched apolipoprotein e-/- mice. CONCLUSIONS Stat1 regulates CD36 expression and foam cell formation in macrophages in vitro; the Stat1 regulation of foam cell formation requires CD36. The regulation of CD36 expression by Stat1 may be important in other pathophysiological CD36-dependent events. Stat1 deficiency reduces atherosclerosis in an apolipoprotein e-/- atherosclerosis-susceptible bone marrow transplantation model.
Collapse
Affiliation(s)
- Sudesh Agrawal
- Department of Cell Biology, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
31
|
Li BL, Chang TY, Chen J, Chang CCY, Zhao XN. Human ACAT1 gene expression and its involvement in the development of atherosclerosis. Future Cardiol 2006; 2:93-9. [DOI: 10.2217/14796678.2.1.93] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis is caused by a series of pathologic changes at the cellular level, with formation of macrophage-derived foam cells occurring at an early stage. Most of the cholesteryl esters in macrophage foam cells are produced by the enzyme acyl-coenzyme A:cholesterol acyltransferase (ACAT). Two ACAT genes, Acat1 and Acat2, exist in mammals. In the monocyte–macrophages, ACAT1 is the major isoenzyme and is a drug target for atherosclerosis treatment. Various proatherogenic stimuli, including interferon-γ and dexamethasone, cause upregulation of human Acat1 expression in macrophages. Thus, it should be possible to find antagonist(s) to downregulate human Acat1 expression. A greater understanding of human Acat1 expression may provide scientists with opportunities for novel therapeutic approaches to combat atherosclerosis.
Collapse
Affiliation(s)
- Bo-Liang Li
- State Key Laboratory of Molecular Biology, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ta-Yuan Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755 USA
| | - Jia Chen
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Catherine CY Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755, USA
| | - Xiao-Nan Zhao
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
32
|
Abstract
Reverse cholesterol transport (RCT) is a pathway by which accumulated cholesterol is transported from the vessel wall to the liver for excretion, thus preventing atherosclerosis. Major constituents of RCT include acceptors such as high-density lipoprotein (HDL) and apolipoprotein A-I (apoA-I), and enzymes such as lecithin:cholesterol acyltransferase (LCAT), phospholipid transfer protein (PLTP), hepatic lipase (HL) and cholesterol ester transfer protein (CETP). A critical part of RCT is cholesterol efflux, in which accumulated cholesterol is removed from macrophages in the subintima of the vessel wall by ATP-binding membrane cassette transporter A1 (ABCA1) or by other mechanisms, including passive diffusion, scavenger receptor B1 (SR-B1), caveolins and sterol 27-hydroxylase, and collected by HDL and apoA-I. Esterified cholesterol in the HDL is then delivered to the liver for excretion. In patients with mutated ABCA1 genes, RCT and cholesterol efflux are impaired and atherosclerosis is increased. In studies with transgenic mice, disruption of ABCA1 genes can induce atherosclerosis. Levels of HDL are inversely correlated with incidences of cardiovascular disease. Supplementation with HDL or apoA-I can reverse atherosclerosis by accelerating RCT and cholesterol efflux. On the other hand, pro-inflammatory factors such as interferon-gamma (IFN-gamma), endotoxin, tumour necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1beta), can be atherogenic by impairing RCT and cholesterol efflux, according to in vitro studies. RCT and cholesterol efflux play a major role in anti-atherogenesis, and modification of these processes may provide new therapeutic approaches to cardiovascular disease. Further research on new modifying factors for RCT and cholesterol efflux is warranted.
Collapse
Affiliation(s)
- R Ohashi
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston 77030, USA
| | | | | | | | | |
Collapse
|
33
|
Daugherty A, Webb NR, Rateri DL, King VL. Thematic review series: The Immune System and Atherogenesis. Cytokine regulation of macrophage functions in atherogenesis. J Lipid Res 2005; 46:1812-22. [PMID: 15995168 DOI: 10.1194/jlr.r500009-jlr200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This review will focus on the role of cytokines in the behavior of macrophages, a prominent cell type of atherosclerotic lesions. Once these macrophages have immigrated into the vessel wall, they propagate the development of atherosclerosis by modifying lipoproteins, accumulating intracellular lipids, remodeling the extracellular environment, and promoting local coagulation. The numerous cytokines that have been detected in atherosclerosis, combined with the expression of large numbers of cytokine receptors on macrophages, are consistent with this axis being an important contributor to lesion development. Given the vast literature on cytokine-macrophage interactions, this review will be selective, with an emphasis on the major cytokines that have been detected in atherosclerotic lesions and their effects on properties that are relevant to lesion formation and maturation. There will be an emphasis on the role of cytokines in regulating lipid metabolism by macrophages. We will provide an overview of the major findings in cell culture and then put these in the context of in vivo studies.
Collapse
Affiliation(s)
- Alan Daugherty
- Cardiovascular Research Center, Gill Heart Institute, University of Kentucky, Lexington, KY, USA.
| | | | | | | |
Collapse
|
34
|
Alfaro Leon ML, Evans GF, Farmen MW, Zuckerman SH. Post-transcriptional regulation of macrophage ABCA1, an early response gene to IFN-γ. Biochem Biophys Res Commun 2005; 333:596-602. [PMID: 15946645 DOI: 10.1016/j.bbrc.2005.05.112] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 05/23/2005] [Indexed: 10/25/2022]
Abstract
Interferon-gamma (IFN-gamma) down-regulates receptors associated with reverse cholesterol transport including ABCA1. In the present study, the kinetics and mechanism of ABCA1 down-regulation were determined in mouse peritoneal macrophages. IFN-gamma decreased ABCA1 mRNA 1h following IFN-gamma addition and was maximally reduced by 3h. Down-regulation was protein synthesis dependent and involved post-transcriptional processes. ABCA1 message had a T(1/2) of 115 min in actinomycin treated cells that was reduced to a T(1/2) of 37 min by IFN-gamma. The decrease in message stability was also associated with a rapid loss of ABCA1 protein, significant 3h following IFN-gamma addition. The kinetics of ABCA1 message and protein decrease was consistent with the early IFN-gamma-induced changes in Stat1 phosphorylation and nuclear translocation observed in these cells. Therefore, ABCA1 can be considered as an early response gene to macrophage activation by IFN-gamma with down-regulation occurring by message destabilization.
Collapse
|
35
|
Koizumi A, Mizukami H, Inoue M. pX Gene Causes Hypercholesterolemia in Hypercholesterolemia-Resistant BALB/c Mice. Biol Pharm Bull 2005; 28:1731-5. [PMID: 16141549 DOI: 10.1248/bpb.28.1731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the high incidence of atherosclerosis in the patients affected with rheumatoid arthritis, we examined the effect of feeding a cholesterol-enriched diet on the development of hypercholesterolemia in pX transgenic mice, which spontaneously develop chronic inflammatory arthritis. Cholesterol feeding to pX transgenic mice induced a striking elevation in serum total cholesterol (ca. 500 mg/dl) compared with their littermates, BALB/c mice used as controls. The pX transgenic mice exhibited elevated mRNA levels of ACAT1, and ABCG5 in the small intestine compared with their littermates, and furthermore, apoA1, ABCA1, ABCG5, ACAT1, and ACAT2 mRNAs were induced more easily by a cholesterol-enriched diet in pX transgenic mice than their littermates. As ACAT1 mRNA in the small intestine is known not to be induced by feeding a cholesterol-enriched diet, a possibility was inferred that interferon-gamma induced by Tax, a pX gene product, might play an important role in the induction of ACAT1 mRNA and the following hypercholesterolemia. These findings suggest that pX gene plays an important role in inducing hypercholesterolemia in BALB/c mice, which are genetically less susceptible to hypercholesterolemia and atherosclerosis and that RA patients carrying HTLV-1 virus have a predilection for hypercholesterolemia, a main risk factor for cardiovascular diseases.
Collapse
Affiliation(s)
- Ayano Koizumi
- Laboratory of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | | | | |
Collapse
|
36
|
Yang L, Yang JB, Chen J, Yu GY, Zhou P, Lei L, Wang ZZ, Cy Chang C, Yang XY, Chang TY, Li BL. Enhancement of human ACAT1 gene expression to promote the macrophage-derived foam cell formation by dexamethasone. Cell Res 2004; 14:315-23. [PMID: 15353128 DOI: 10.1038/sj.cr.7290231] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In macrophages, the accumulation of cholesteryl esters synthesized by the activated acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT1) results in the foam cell formation, a hallmark of early atherosclerotic lesions. In this study, with the treatment of a glucocorticoid hormone dexamethasone (Dex), lipid staining results clearly showed the large accumulation of lipid droplets containing cholesteryl esters in THP-1-derived macrophages exposed to lower concentration of the oxidized low-density lipoprotein (ox-LDL). More notably, when treated together with specific anti-ACAT inhibitors, the abundant cholesteryl ester accumulation was markedly diminished in THP-1-derived macrophages, confirming that ACAT is the key enzyme responsible for intracellular cholesteryl ester synthesis. RT-PCR and Western blot results indicated that Dex caused up-regulation of human ACAT1 expression at both the mRNA and protein levels in THP-1 and THP-1-derived macrophages. The luciferase activity assay demonstrated that Dex could enhance the activity of human ACAT1 gene P1 promoter, a major factor leading to the ACAT1 activation, in a cell-specific manner. Further experimental evidences showed that a glucocorticoid response element (GRE) located within human ACAT1 gene P1 promoter to response to the elevation of human ACAT1 gene expression by Dex could be functionally bound with glucocorticoid receptor (GR) proteins. These data supported the hypothesis that the clinical treatment with Dex, which increased the incidence of atherosclerosis, may in part due to enhancing the ACAT1 expression to promote the accumulation of cholesteryl esters during the macrophage-derived foam cell formation, an early stage of atherosclerosis.
Collapse
Affiliation(s)
- Li Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Stolzing A, Grune T. Neuronal apoptotic bodies: phagocytosis and degradation by primary microglial cells. FASEB J 2004; 18:743-5. [PMID: 14766802 DOI: 10.1096/fj.03-0374fje] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neuronal loss via apoptosis is a key element in numerous neurodegenerative diseases. To avoid accumulation of apoptotic material, the remains of apoptotic cells should be degraded. It was suggested that microglial cells are phagocytosing and degrading apoptotic material. There is only limited information available concerning the fate of the remains of apoptotic neurons. In this study, we investigated the ability of microglial cells to take up and degrade neuronal apoptotic material. We isolated primary microglial cells and used apoptotic bodies of apoptotic neuron-like PC12 cells as a substrate. The apoptotic material was taken up and degraded within the microglial cells. The uptake is clearly activation dependent. We were able to demonstrate that the CD36 scavenger receptor is involved in the uptake of the apoptotic material via competition studies, antibody blockage, and use of a CD36 mutant rat strain. Blockage of other uptake mechanisms was also able to inhibit the uptake to some extent. Furthermore, we were able to demonstrate the role of the microglial lysosomal and proteasomal pathways in the degradation of proteins originating from apoptotic bodies.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Neuroscience Research Center, Medical Faculty (Charité), Humboldt University, Berlin, Germany
| | | |
Collapse
|
38
|
Abstract
Atherosclerosis is an inflammatory disease. T lymphocytes, occurring concomitantly with macrophages, are found in atherosclerotic lesions with substantial numbers in all stages. Most of the T cells in the lesions are CD4(+) T cells. The finding of activated T cells and macrophages in lesions and cloning of T cells specific for modified low-density lipoproteins from lesions suggest that a cell-mediated immune reaction is taking place in atherosclerosis. This review provides an overview of our current understanding of the roles of CD4(+) T cell subpopulations in atherosclerosis.
Collapse
Affiliation(s)
- Xinghua Zhou
- Center for Molecular Medicine L8:03 and Department of Medicine, Karolinska Hospital, Karolinska Institutet, 17176, Stockholm, Sweden.
| |
Collapse
|
39
|
Chinetti G, Lestavel S, Fruchart JC, Clavey V, Staels B. Peroxisome proliferator-activated receptor alpha reduces cholesterol esterification in macrophages. Circ Res 2003; 92:212-7. [PMID: 12574149 DOI: 10.1161/01.res.0000053386.46813.e9] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARalpha) is a nuclear receptor activated by fatty acid derivatives and hypolipidemic drugs of the fibrate class. PPARalpha is expressed in monocytes, macrophages, and foam cells, suggesting a role for this receptor in macrophage lipid homeostasis with consequences for atherosclerosis development. Recently, it was shown that PPARalpha activation promotes cholesterol efflux from macrophages via induction of the ABCA1 pathway. In the present study, the influence of PPARalpha activators on intracellular cholesterol homeostasis was investigated. In human macrophages and foam cells, treatment with fibrates, synthetic PPARalpha activators, led to a decrease in the cholesteryl ester (CE):free cholesterol (FC) ratio. In these cells, PPARalpha activation reduced cholesterol esterification rates and Acyl-CoA:cholesterol acyltransferase-1 (ACAT1) activity. However, PPARalpha activation did not alter ACAT1 gene expression, whereas mRNA levels of carnitine palmitoyltransferase type 1 (CPT-1), a key enzyme in mitochondrial fatty acid catabolism, were induced. Finally, PPARalpha activation blocked CE formation induced by TNF-alpha, possibly due to the inhibition of neutral sphingomyelinase activation by TNF-alpha. In conclusion, our results identify a role for PPARalpha in the control of cholesterol esterification in macrophages, resulting in an enhanced availability of FC for efflux through the ABCA1 pathway.
Collapse
Affiliation(s)
- G Chinetti
- UR 545 INSERM, Institut Pasteur de Lille and Université de Lille 2, Lille, France.
| | | | | | | | | |
Collapse
|
40
|
Sun YH, den Hartigh AB, Santos RL, Adams LG, Tsolis RM. virB-Mediated survival of Brucella abortus in mice and macrophages is independent of a functional inducible nitric oxide synthase or NADPH oxidase in macrophages. Infect Immun 2002; 70:4826-32. [PMID: 12183526 PMCID: PMC128286 DOI: 10.1128/iai.70.9.4826-4832.2002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2002] [Revised: 05/28/2002] [Accepted: 06/06/2002] [Indexed: 11/20/2022] Open
Abstract
The Brucella abortus virB locus is required for establishing chronic infection in the mouse. Using in vitro and in vivo models, we investigated whether virB is involved in evasion of the bactericidal activity of NADPH oxidase and the inducible nitric oxide synthase (iNOS) in macrophages. Elimination of NADPH oxidase or iNOS activity in macrophages in vitro increased recovery of wild-type B. abortus but not recovery of a virB mutant. In mice lacking either NADPH oxidase or iNOS, however, B. abortus infected and persisted to the same extent as it did in congenic C57BL/6 mice up until 60 days postinfection, suggesting that these host defense mechanisms are not critical for limiting bacterial growth in the mouse. A virB mutant did not exhibit increased survival in either of the knockout mouse strains, indicating that this locus does not contribute to evasion of nitrosative or oxidative killing mechanisms in vivo.
Collapse
Affiliation(s)
- Yao-Hui Sun
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station, USA
| | | | | | | | | |
Collapse
|
41
|
Zuckerman SH, Kauffman RF, Evans GF. Peroxisome proliferator-activated receptor alpha,gamma coagonist LY465608 inhibits macrophage activation and atherosclerosis in apolipoprotein E knockout mice. Lipids 2002; 37:487-94. [PMID: 12056591 DOI: 10.1007/s11745-002-0922-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The apolipoprotein E (apoE) knockout mouse has provided an approach to the investigation of the effect of both cellular and humoral processes on atherosclerotic lesion progression. In the present study, pharmacologic modulation of both interferon gamma (IFNgamma)-inducible macrophage effector functions, and atherosclerotic lesions in the apoE knockout mouse were investigated using the peroxisome proliferator-activated receptor (PPAR) alpha,gamma coagonist LY465608. LY465608 inhibited, in a concentration-dependent manner, IFNgamma induction of both nitric oxide synthesis and the beta 2 integrin CD11a in elicited peritoneal macrophages from apoE knockout mice. Similar effects were observed ex vivo following 10 d of treating mice with 10 mg/kg of LY465608. Treatment of apoE knockout mice for 18 wk with LY465608 resulted in a statistically significant 2.5-fold reduction in atherosclerotic lesion area in en face aorta preparations. These effects were apparent in the absence of any reduction in total serum cholesterol or in lipoprotein distribution. Finally, treatment of apoF knockout mice with established atherosclerotic disease resulted in a modest but not statistically significant decrease in aortic lesional surface area. These results demonstrate the utility of PPAR coagonists in reducing the progression of the atherosclerotic lesion.
Collapse
Affiliation(s)
- Steven H Zuckerman
- Division of Cardiovascular Research, Lilly Research Laboratories, Indianapolis, Indiana 46285, USA.
| | | | | |
Collapse
|
42
|
Wang XQ, Panousis CG, Alfaro ML, Evans GF, Zuckerman SH. Interferon-gamma-mediated downregulation of cholesterol efflux and ABC1 expression is by the Stat1 pathway. Arterioscler Thromb Vasc Biol 2002; 22:e5-9. [PMID: 12006410 DOI: 10.1161/01.atv.0000018287.03856.dd] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The pathological role of interferon-gamma (IFN-gamma) in atherosclerosis is mediated through effects on macrophages, foam cells, and other vascular cells. Recently, we reported that ATP-binding cassette transporter 1(ABC1) message and protein levels were decreased 3- to 4-fold in foam cells by IFN-gamma. In the present study, the pathway by which IFN-gamma inhibited ABC1 expression was investigated with signal transducers and activators of transcription (Stat1) knockout mice. IFN-gamma-stimulated, wild-type, macrophage-derived foam cells, as previously reported, exhibited a decrease in cholesterol efflux and ABC1 expression as well as an increase in acyl coenzyme A:cholesterol-O-acyltransferase activity. However, IFN-gamma treatment of foam cells from Stat1 knockout mice failed to demonstrate reductions in efflux or ABC1 expression at the message or protein levels, nor were there any increases in acyl coenzyme A:cholesterol-O-acyltransferase activity. However, ABC1 mRNA expression in macrophages from Stat1 knockout mice could still be demonstrated to be increased by lipid loading with acetylated low density lipoprotein. Finally, Stat1-independent gene activation by IFN-gamma was intact in the Stat1 KO macrophages, inasmuch as IFN-gamma was shown to stimulate increases in interleukin-6 production in the Stat1 KO macrophages that were comparable to those observed in the wild-type macrophages. Therefore, Stat1 signaling is necessary and sufficient for the inhibitory effects of IFN-gamma on cholesterol efflux and ABC1 expression.
Collapse
Affiliation(s)
- Xue-Qing Wang
- Division of Cardiovascular Research, Lilly Research Labs, Indianapolis, Ind 46285, USA
| | | | | | | | | |
Collapse
|
43
|
Abstract
Although the concept that inflammation plays a role in the biology of atherosclerosis is now well accepted, the basic feature of the arterial lesion remains the accumulation of clusters of foam cells. These clusters are the consequence of the enhanced recruitment of monocytes in the vessel wall induced by the hyperlipidemia and of the disproportionate accumulation of lipids in the cytoplasm of macrophages deriving from monocytes. Ultimately, every molecular force and pathway with modulating activity over the developing lesion will have to act on a convergence point with factors regulating cholesterol balance in the macrophage. Consistent with this view is the recent report that cytokines, such as tumor necrosis factor-alpha, can influence the expression of the scavenger receptor, whereas interferon-gamma can inhibit adenosine triphosphate-binding cassette transporter-1, the main effector of cholesterol efflux in the peripheral cell. Conversely, recent data have shown that primary alterations in macrophage cholesterol balance, such as those produced by the total absence of acylcoenzyme A:cholesterol acyltransferase-1, may determine local changes compatible with the activation of inflammatory pathways. In this brief review, we discuss some of the convergence points between inflammation and cholesterol balance, and we highlight the additional therapeutic targets suggested by these new developments in vascular biology.
Collapse
Affiliation(s)
- S Fazio
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6300, USA.
| | | |
Collapse
|
44
|
Yang JB, Duan ZJ, Yao W, Lee O, Yang L, Yang XY, Sun X, Chang CC, Chang TY, Li BL. Synergistic transcriptional activation of human Acyl-coenzyme A: cholesterol acyltransterase-1 gene by interferon-gamma and all-trans-retinoic acid THP-1 cells. J Biol Chem 2001; 276:20989-98. [PMID: 11399774 DOI: 10.1074/jbc.m011488200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acyl-coenzyme A:cholesterol acyltransferase (ACAT) is an intracellular enzyme involved in cellular cholesterol homeostasis and in atherosclerotic foam cell formation. Human ACAT-1 gene contains two promoters (P1 and P7), each located in a different chromosome (1 and 7) (Li, B. L., Li, X. L., Duan, Z. J., Lee, O., Lin, S., Ma, Z. M., Chang, C. C., Yang, X. Y., Park, J. P., Mohandas, T. K., Noll, W., Chan, L., and Chang, T. Y. (1999) J. Biol Chem. 274, 11060-11071). Interferon-gamma (IFN-gamma), a cytokine that exerts many pro-atherosclerotic effects in vivo, causes up-regulation of ACAT-1 mRNA in human blood monocyte-derived macrophages and macrophage-like cells but not in other cell types. To examine the molecular nature of this observation, we identified within the ACAT-1 P1 promoter a 159-base pair core region. This region contains 4 Sp1 elements and an IFN-gamma activated sequence (GAS) that overlaps with the second Sp1 element. In the monocytic cell line THP-1 cell, the combination of IFN-gamma and all-trans-retinoic acid (a known differentiation agent) enhances the ACAT-1 P1 promoter but not the P7 promoter. Additional experiments showed that all-trans-retinoic acid causes large induction of the transcription factor STAT1, while IFN-gamma causes activation of STAT1 such that it binds to the GAS/Sp1 site in the ACAT-1 P1 promoter. Our work provides a molecular mechanism to account for the effect of IFN-gamma in causing transcriptional activation of ACAT-1 in macrophage-like cells.
Collapse
Affiliation(s)
- J B Yang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Acyl-coenzyme A:cholesterol acyltransferase (ACAT) is an intracellular enzyme that produces cholesteryl esters in various tissues. In mammals, two ACAT genes (ACAT1 and ACAT2) have been identified. Together, these two enzymes are involved in storing cholesteryl esters as lipid droplets, in macrophage foam-cell formation, in absorbing dietary cholesterol, and in supplying cholesteryl esters as part of the core lipid for lipoprotein synthesis and assembly. The key difference in tissue distribution of ACAT1 and ACAT2 between humans, mice and monkeys is that, in adult human liver (including hepatocytes and bile duct cells), the major enzyme is ACAT1, rather than ACAT2. There is compelling evidence implicating a role for ACAT1 in macrophage foam-cell formation, and for ACAT2 in intestinal cholesterol absorption. However, further studies at the biochemical and cell biological levels are needed in order to clarify the functional roles of ACAT1 and ACAT2 in the VLDL or chylomicron synthesis/assembly process.
Collapse
Affiliation(s)
- T Y Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Panousis CG, Evans G, Zuckerman SH. TGF-β increases cholesterol efflux and ABC-1 expression in macrophage-derived foam cells: opposing the effects of IFN-γ. J Lipid Res 2001. [DOI: 10.1016/s0022-2275(20)31648-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
47
|
Zuckerman SH, Panousis C, Evans G. TGF-beta reduced binding of high-density lipoproteins in murine macrophages and macrophage-derived foam cells. Atherosclerosis 2001; 155:79-85. [PMID: 11223429 DOI: 10.1016/s0021-9150(00)00540-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The expression of macrophage scavenger receptors is regulated by intracellular cholesterol levels, as well as by cytokines affecting macrophage effector functions. CD36, a member of the type B scavenger receptor family, will bind a variety of nonlipoprotein and lipoprotein ligands including high-density lipoprotein (HDL). Transforming growth factor-beta (TGF-beta) has been demonstrated to modulate macrophage effector functions and is present within atherosclerotic lesions. In the present study, the effect of TGF-beta on HDL binding by both macrophages and macrophage-derived foam cells was evaluated. TGF-beta, in a dose-dependent manner, reduced the binding of flurochrome-labeled HDL to both macrophages and foam cells. These effects were observed in macrophages derived from nonatherosclerotic (BALB/c) as well as from macrophages obtained from both apolipoprotein E and low-density lipoprotein receptor knockout mice. The decrease in HDL binding was consistent with a significant reduction in CD36 message levels. The effect of TGF-beta on type B scavenger receptor expression was not limited to CD36 as SR-BI message was also downregulated, although the effect was more modest. A similar reduction in HDL binding and CD36 message was also observed with the immunosuppressive glucocorticoid dexamethasone. These results suggest that within the microenvironment of an atherosclerotic lesion, TGF-beta and other agents that inhibit macrophage inflammatory responses may impact lesion progression through mechanisms that include the modulation of HDL-foam cell interactions.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/genetics
- Arteriosclerosis/metabolism
- Blotting, Northern
- CD36 Antigens/metabolism
- Cells, Cultured
- Dexamethasone/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Flow Cytometry
- Foam Cells/metabolism
- Immunosuppressive Agents
- Lipoproteins, HDL/metabolism
- Macrophages, Peritoneal/metabolism
- Membrane Proteins
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Receptors, Immunologic/metabolism
- Receptors, LDL/genetics
- Receptors, Lipoprotein
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- S H Zuckerman
- Division of Cardiovascular Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA.
| | | | | |
Collapse
|
48
|
Maung KK, Miyazaki A, Nomiyama H, Chang CC, Chang TY, Horiuchi S. Induction of acyl-coenzyme A:cholesterol acyltransferase-1 by 1,25-dihydroxyvitamin D3 or 9-cis-retinoic acid in undifferentiated THP-1 cells. J Lipid Res 2001. [DOI: 10.1016/s0022-2275(20)31677-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
49
|
Chen W, Silver DL, Smith JD, Tall AR. Scavenger receptor-BI inhibits ATP-binding cassette transporter 1- mediated cholesterol efflux in macrophages. J Biol Chem 2000; 275:30794-800. [PMID: 10896940 DOI: 10.1074/jbc.m004552200] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Scavenger receptor BI (SR-BI) facilitates the efflux of cellular cholesterol to plasma high density lipoprotein (HDL). Recently, the ATP-binding cassette transporter 1 (ABC1) was identified as a key mediator of cholesterol efflux to apolipoproteins and HDL. The goal of the present study was to determine a possible interaction between the SR-BI and ABC1 cholesterol efflux pathways in macrophages. Free cholesterol efflux to HDL was increased ( approximately 2.2-fold) in SR-BI transfected RAW macrophages in association with increased SR-BI protein levels. Treatment of macrophages with 8-bromo-cAMP (cAMP) resulted in a 4.1-fold increase in ABC1 mRNA level and also increased cholesterol efflux to HDL (2.2-fold) and apoA-I (5.5-fold). However, in SR-BI transfected RAW cells, cAMP treatment produced a much smaller increment in cholesterol efflux to HDL (1.1-fold) or apoA-I (3.3-fold) compared with control cells. In macrophages loaded with cholesterol by acetyl-LDL treatment, SR-BI overexpression did not increase cholesterol efflux to HDL but did inhibit cAMP-mediated cholesterol efflux to apoA-I or HDL. SR-BI neutralizing antibody led to a dose- and time-dependent increase of cAMP-mediated cholesterol efflux in both SR-BI transfected and control cells, indicating that SR-BI inhibits ABC1-mediated cholesterol efflux even at low SR-BI expression level. Transfection of a murine ABC1 cDNA into 293 cells led to a 2.3-fold increase of cholesterol efflux to apoA-I, whereas co-transfection of SR-BI with ABC1 blocked this increase in cholesterol efflux. SR-BI and ABC1 appear to have distinct and competing roles in mediating cholesterol flux between HDL and macrophages. In nonpolarized cells, SR-BI promotes the reuptake of cholesterol actively effluxed by ABC1, creating a futile cycle.
Collapse
Affiliation(s)
- W Chen
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, New York 10032, USA
| | | | | | | |
Collapse
|
50
|
Ares MP. Bimonthly update: lipidology. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol 2000; 11:563-5. [PMID: 11048900 DOI: 10.1097/00041433-200010000-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|