251
|
Lathe R, Sapronova A, Kotelevtsev Y. Atherosclerosis and Alzheimer--diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr 2014; 14:36. [PMID: 24656052 PMCID: PMC3994432 DOI: 10.1186/1471-2318-14-36] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/26/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Aging is accompanied by increasing vulnerability to pathologies such as atherosclerosis (ATH) and Alzheimer disease (AD). Are these different pathologies, or different presentations with a similar underlying pathoetiology? DISCUSSION Both ATH and AD involve inflammation, macrophage infiltration, and occlusion of the vasculature. Allelic variants in common genes including APOE predispose to both diseases. In both there is strong evidence of disease association with viral and bacterial pathogens including herpes simplex and Chlamydophila. Furthermore, ablation of components of the immune system (or of bone marrow-derived macrophages alone) in animal models restricts disease development in both cases, arguing that both are accentuated by inflammatory/immune pathways. We discuss that amyloid β, a distinguishing feature of AD, also plays a key role in ATH. Several drugs, at least in mouse models, are effective in preventing the development of both ATH and AD. Given similar age-dependence, genetic underpinnings, involvement of the vasculature, association with infection, Aβ involvement, the central role of macrophages, and drug overlap, we conclude that the two conditions reflect different manifestations of a common pathoetiology. MECHANISM Infection and inflammation selectively induce the expression of cholesterol 25-hydroxylase (CH25H). Acutely, the production of 'immunosterol' 25-hydroxycholesterol (25OHC) defends against enveloped viruses. We present evidence that chronic macrophage CH25H upregulation leads to catalyzed esterification of sterols via 25OHC-driven allosteric activation of ACAT (acyl-CoA cholesterol acyltransferase/SOAT), intracellular accumulation of cholesteryl esters and lipid droplets, vascular occlusion, and overt disease. SUMMARY We postulate that AD and ATH are both caused by chronic immunologic challenge that induces CH25H expression and protection against particular infectious agents, but at the expense of longer-term pathology.
Collapse
Affiliation(s)
- Richard Lathe
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Pieta Research, PO Box 27069, Edinburgh EH10 5YW, UK
| | - Alexandra Sapronova
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Optical Research Group, Laboratory of Evolutionary Biophysics of Development, Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Yuri Kotelevtsev
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Biomedical Centre for Research Education and Innovation (CREI), Skolkovo Institute of Science and Technology, Skolkovo 143025, Russia
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Little France, Edinburgh EH16 4TJ, UK
| |
Collapse
|
252
|
Yilmaz A, Jennbacken K, Fogelstrand L. Reduced IgM levels and elevated IgG levels against oxidized low-density lipoproteins in HIV-1 infection. BMC Infect Dis 2014; 14:143. [PMID: 24636004 PMCID: PMC3995360 DOI: 10.1186/1471-2334-14-143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/12/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Chronic HIV infection is associated with increased risk of cardiovascular disease caused by atherosclerosis. Oxidized forms of low-density lipoprotein (LDL) are present in atherosclerotic lesions and constitute major epitopes for natural antibodies. IgM has been shown to be protective against atherosclerosis, whereas the role of corresponding IgG is less clear. The objective of this study was to determine if HIV + individuals have disturbed levels of IgM and IgG directed against oxidized forms of LDL as compared to HIV- individuals. METHODS Ninety-one HIV + patients and 92 HIV- controls were included in this retrospective study. Circulating levels of IgG and IgM directed against two forms of oxidized LDL; copper oxidized (OxLDL) and malondialdehyde modified (MDA-LDL), total IgM and IgG, C-reactive protein (CRP), soluble CD14, and apolipoproteins A1 and B were determined. RESULTS HIV + individuals had slightly lower levels of IgM against MDA-LDL and higher levels of IgG against MDA-LDL, OxLDL, and total IgG, than HIV- controls. Anti-MDA-LDL and Anti-OxLDL IgG displayed a positive correlation with viral load and a negative correlation with the CD4+ T-cell count. HIV + individuals also displayed elevated CRP and soluble CD14 levels compared to HIV- individuals, but there were no correlations between CRP or soluble CD14 and specific antibodies. CONCLUSIONS HIV infection is associated with higher levels of IgG including specific IgG against oxidized forms of LDL, and lower IgM against the same epitope. In addition to dyslipidemia, immune activation, HIV-replication and an accumulation of risk factors for atherosclerosis, this adverse antibody profile may be of major importance for the increased risk of cardiovascular disease in HIV + individuals.
Collapse
Affiliation(s)
- Aylin Yilmaz
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Jennbacken
- Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linda Fogelstrand
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, all at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
253
|
Blanco-Colio LM. TWEAK/Fn14 Axis: A Promising Target for the Treatment of Cardiovascular Diseases. Front Immunol 2014; 5:3. [PMID: 24478772 PMCID: PMC3895871 DOI: 10.3389/fimmu.2014.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/03/2014] [Indexed: 11/30/2022] Open
Abstract
Cardiovascular diseases (CVD) are the first cause of mortality in Western countries. CVD include several pathologies such as coronary heart disease, stroke or cerebrovascular accident, congestive heart failure, peripheral arterial disease, and aortic aneurysm, among others. Interaction between members of the tumor necrosis factor (TNF) superfamily and their receptors elicits several biological actions that could participate in CVD. TNF-like weak inducer of apoptosis (TWEAK) and its functional receptor and fibroblast growth factor-inducible molecule 14 (Fn14) are two proteins belonging to the TNF superfamily that activate NF-κB by both canonical and non-canonical pathways and regulate several cell functions such as proliferation, migration, differentiation, cell death, inflammation, and angiogenesis. TWEAK/Fn14 axis plays a beneficial role in tissue repair after acute injury. However, persistent TWEAK/Fn14 activation mediated by blocking experiments or overexpression experiments in animal models has shown an important role of this axis in the pathological remodeling underlying CVD. In this review, we summarize the role of TWEAK/Fn14 pathway in the development of CVD, focusing on atherosclerosis and stroke and the molecular mechanisms by which TWEAK/Fn14 interaction participates in these pathologies. We also review the role of the soluble form of TWEAK as a biomarker for the diagnosis and prognosis of CVD. Finally, we highlight the results obtained with other members of the TNF superfamily that also activate canonical and non-canonical NF-κB pathway.
Collapse
|
254
|
Affiliation(s)
- Gabrielle Fredman
- From the Institute of Molecular Cardiology (M.S.), Diabetes and Obesity Center (M.S.), and Department of Microbiology and Immunology (M.S.), University of Louisville, Louisville, KY; and Department of Medicine, Columbia University, New York, NY (G.F.)
| | - Matthew Spite
- From the Institute of Molecular Cardiology (M.S.), Diabetes and Obesity Center (M.S.), and Department of Microbiology and Immunology (M.S.), University of Louisville, Louisville, KY; and Department of Medicine, Columbia University, New York, NY (G.F.)
| |
Collapse
|
255
|
Morris-Rosenfeld S, Lipinski MJ, McNamara CA. Understanding the role of B cells in atherosclerosis: potential clinical implications. Expert Rev Clin Immunol 2013; 10:77-89. [PMID: 24308836 DOI: 10.1586/1744666x.2014.857602] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a progressive inflammatory disease of the medium to large arteries that is the largest contributor to cardiovascular disease. B-cell subsets have been shown in animal models of atherosclerosis to have both atherogenic and atheroprotective properties. In this review, we highlight the research that developed our understanding of the role of B cells in atherosclerosis both in humans and mice. From this we discuss the potential clinical impact B cells could have both as diagnostic biomarkers and as targets for immunotherapy. Finally, we recognize the inherent difficulty in translating findings from animal models into humans given the differences in both cardiovascular disease and the immune system between mice and humans, making the case for greater efforts at addressing the role of B cells in human atherosclerosis.
Collapse
Affiliation(s)
- Samuel Morris-Rosenfeld
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA and Department of Medicine, Cardiovascular Division at the University of Virginia, Charlottesville, VA, USA
| | | | | |
Collapse
|
256
|
Smeets E, Meiler S, Lutgens E. Lymphocytic tumor necrosis factor receptor superfamily co-stimulatory molecules in the pathogenesis of atherosclerosis. Curr Opin Lipidol 2013; 24:518-24. [PMID: 24184937 DOI: 10.1097/mol.0000000000000025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW The role of lymphocytes in the chronic inflammatory disease atherosclerosis has emerged over the past decade. Co-stimulatory molecules of the heterogeneous tumor necrosis factor receptor superfamily play a pivotal role in lymphocyte activation, proliferation and differentiation. Here we describe the immune modulatory properties and mechanisms of four tumor necrosis factor receptor superfamily members in atherosclerosis. RECENT FINDINGS CD40/CD40L, OX40L/OX40, CD70/CD27 and CD137/CD137L are present in human atherosclerotic plaques and have shown strong immune modulatory functions in atherosclerosis, resulting in either atherogenic or atheroprotective effects in mouse models of atherosclerosis. SUMMARY Insight into the immune modulatory mechanisms of co-stimulatory interactions in atherosclerosis can contribute to clinical exploitation of these interactions in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Esther Smeets
- aDepartment of Medical Biochemistry, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands bInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, ,Germany
| | | | | |
Collapse
|
257
|
Mantani PT, Ljungcrantz I, Andersson L, Alm R, Hedblad B, Björkbacka H, Nilsson J, Fredrikson GN. Circulating CD40+ and CD86+ B cell subsets demonstrate opposing associations with risk of stroke. Arterioscler Thromb Vasc Biol 2013; 34:211-8. [PMID: 24202305 DOI: 10.1161/atvbaha.113.302667] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Accumulating evidence shows that immune cells play an important role in atherosclerosis. Most attention has focused on the role of different T cell subsets, whereas the possible involvement of B cells has been less studied. In this study, we assessed the association of 2 different B cell subsets, CD19(+)CD40(+) and CD19(+)CD86(+) B cells, with risk for development of acute cardiovascular events. APPROACH AND RESULTS The prospective study included 700 subjects randomly selected from the cardiovascular cohort of the Malmö Diet and Cancer study. Mononuclear leukocytes, stored at -140(○)C at the baseline investigation in 1991-1994, were thawed and B cell subsets analyzed by flow cytometry. Cytokine release from CD3/CD28-stimulated mononuclear leukocytes was measured with multiplex ELISA. Baseline carotid intima-media thickness and stenosis were assessed by ultrasonography, and clinical events were monitored through validated national registers during a median/mean follow-up time of 15 years. The subjects in the highest tertile of CD19(+)CD40(+) B cells had a significantly lower risk of incident stroke after adjustment for other risk factors. In contrast, CD19(+)CD86(+) B cells were associated with higher risk for development of a stroke event and increased release of proinflammatory cytokines from mononuclear leukocytes. CONCLUSIONS These observations provide evidence for an involvement of B cells in the incidence of stroke and suggest that both pathogenic and protective B cell subsets exist.
Collapse
Affiliation(s)
- Polyxeni T Mantani
- From the Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden (P.T.M., I.L., L.A., R.A., B.H., H.B., J.N., G.N.F.); and Faculty of Health and Society, Malmö University, Malmö, Sweden (G.N.F.)
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Yu F, Du F, Wang Y, Huang S, Miao R, Major AS, Murphy EA, Fu M, Fan D. Bone marrow deficiency of MCPIP1 results in severe multi-organ inflammation but diminishes atherogenesis in hyperlipidemic mice. PLoS One 2013; 8:e80089. [PMID: 24223214 PMCID: PMC3819309 DOI: 10.1371/journal.pone.0080089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/08/2013] [Indexed: 11/20/2022] Open
Abstract
Objective MCPIP1 is a newly identified protein that profoundly impacts immunity and inflammation. We aim to test if MCPIP1 deficiency in hematopoietic cells results in systemic inflammation and accelerates atherogenesis in mice. Approach and Results After lethally irradiated, LDLR−/− mice were transplanted with bone marrow cells from either wild-type or MCPIP1−/− mice. These chimeric mice were fed a western-type diet for 7 weeks. We found that bone marrow MCPIP1−/− mice displayed a phenotype similar to that of whole body MCPIP1−/− mice, with severe systemic and multi-organ inflammation. However, MCPIP1−/− bone marrow recipients developed >10-fold less atherosclerotic lesions in the proximal aorta than WT bone marrow recipients, and essentially no lesions in en face aorta. The diminishment in atherosclerosis in bone marrow MCPIP1−/− mice may be partially attributed to the slight decrease in their plasma lipids. Flow cytometric analysis of splenocytes showed that bone marrow MCPIP1−/− mice contained reduced numbers of T cells and B cells, but increased numbers of regulatory T cells, Th17 cells, CD11b+/Gr1+ cells and CD11b+/Ly6Clow cells. This overall anti-atherogenic leukocyte profile may also contribute to the reduced atherogenesis. We also examined the cholesterol efflux capability of MCPIP1 deficient macrophages, and found that MCPIP1deficiency increased cholesterol efflux to apoAI and HDL, due to increased protein levels of ABCA1 and ABCG1. Conclusions Hematopoietic deficiency of MCPIP1 resulted in severe systemic and multi-organ inflammation but paradoxically diminished atherogenesis in mice. The reduced atheroegensis may be explained by the decreased plasma cholesterol levels, the anti-atherogenic leukocyte profile, as well as enhanced cholesterol efflux capability. This study suggests that, while atherosclerosis is a chronic inflammatory disease, the mechanisms underlying atherogenesis-associated inflammation in arterial wall versus the inflammation in solid organs may be substantially different.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nutrition and Food Hygiene, the Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Fen Du
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- Department of Biochemistry and molecular Biology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Shengping Huang
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Ruidong Miao
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Amy S. Major
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United State of America
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Mingui Fu
- Shock/Trauma Research Center & Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, Missouri, United States of America
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
259
|
Cardiovascular risk in rheumatoid arthritis: How to lower the risk? Atherosclerosis 2013; 231:163-72. [DOI: 10.1016/j.atherosclerosis.2013.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/30/2013] [Accepted: 09/08/2013] [Indexed: 12/21/2022]
|
260
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
261
|
Winer DA, Winer S, Chng MHY, Shen L, Engleman EG. B Lymphocytes in obesity-related adipose tissue inflammation and insulin resistance. Cell Mol Life Sci 2013; 71:1033-43. [PMID: 24127133 DOI: 10.1007/s00018-013-1486-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/06/2013] [Accepted: 09/26/2013] [Indexed: 12/11/2022]
Abstract
Obesity-related insulin resistance is a chronic inflammatory condition that often gives rise to type 2 diabetes (T2D). Much evidence supports a role for pro-inflammatory T cells and macrophages in promoting local inflammation in tissues such as visceral adipose tissue (VAT) leading to insulin resistance. More recently, B cells have emerged as an additional critical player in orchestrating these processes. B cells infiltrate VAT and display functional and phenotypic changes in response to diet-induced obesity. B cells contribute to insulin resistance by presenting antigens to T cells, secreting inflammatory cytokines, and producing pathogenic antibodies. B cell manipulation represents a novel approach to the treatment of obesity-related insulin resistance and potentially to the prevention of T2D. This review summarizes the roles of B cells in governing VAT inflammation and the mechanisms by which these cells contribute to altered glucose homeostasis in insulin resistance.
Collapse
Affiliation(s)
- Daniel A Winer
- Department of Pathology, Toronto General Hospital, University Health Network, University of Toronto, Eaton Wing, 11E - 424A, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | | | | | | | | |
Collapse
|
262
|
Karper JC, de Jager SCA, Ewing MM, de Vries MR, Bot I, van Santbrink PJ, Redeker A, Mallat Z, Binder CJ, Arens R, Jukema JW, Kuiper J, Quax PHA. An unexpected intriguing effect of Toll-like receptor regulator RP105 (CD180) on atherosclerosis formation with alterations on B-cell activation. Arterioscler Thromb Vasc Biol 2013; 33:2810-7. [PMID: 24115036 DOI: 10.1161/atvbaha.113.301882] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE In atherosclerosis, Toll-like receptors (TLRs) are traditionally linked to effects on tissue macrophages or foam cells. RP105, a structural TLR4 homolog, is an important regulator of TLR signaling. The effects of RP105 on TLR signaling vary for different leukocyte subsets known to be involved in atherosclerosis, making it unique in its role of either suppressing (in myeloid cells) or enhancing (in B cells) TLR-regulated inflammation in different cell types. We aimed to identify a role of TLR accessory molecule RP105 on circulating cells in atherosclerotic plaque formation. APPROACH AND RESULTS Irradiated low density lipoprotein receptor deficient mice received RP105(-/-) or wild-type bone marrow. RP105(-/-) chimeras displayed a 57% reduced plaque burden. Interestingly, total and activated B-cell numbers were significantly reduced in RP105(-/-) chimeras. Activation of B1 B cells was unaltered, suggesting that RP105 deficiency only affected inflammatory B2 B cells. IgM levels were unaltered, but anti-oxidized low-density lipoprotein and anti-malondialdehyde-modified low-density lipoprotein IgG2c antibody levels were significantly lower in RP105(-/-) chimeras, confirming effects on B2 B cells rather than B1 B cells. Moreover, B-cell activating factor expression was reduced in spleens of RP105(-/-) chimeras. CONCLUSIONS RP105 deficiency on circulating cells results in an intriguing unexpected TLR-associated mechanisms that decrease atherosclerotic lesion formation with alterations on proinflammatory B2 B cells.
Collapse
Affiliation(s)
- J C Karper
- From the Department of Surgery (J.C.K., M.M.E., M.R.d.V., J.W.J., P.H.A.Q.), Einthoven Laboratory for Experimental Vascular Medicine (J.C.K., M.M.E., M.R.d.V., P.H.A.Q.), Department of Cardiology (M.M.E., J.W.J.), and Department of Immunohematology and Blood Transfusion (A.R., R.A.), Leiden University Medical Center, Leiden, The Netherlands; Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands (S.C.A.d.J.); Division of Biopharmaceutics, Leiden Amsterdam Center for Drug Research, Leiden University, Leiden, The Netherlands (S.C.A.d.J., I.B., P.J.v.S., J.K.); Inserm U970, Paris Cardiovascular Research Center, Paris, France (Z.M.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (C.J.B.); and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Kummu O, Turunen SP, Wang C, Lehtimäki J, Veneskoski M, Kastarinen H, Koivula MK, Risteli J, Kesäniemi YA, Hörkkö S. Carbamyl adducts on low-density lipoprotein induce IgG response in LDLR-/- mice and bind plasma autoantibodies in humans under enhanced carbamylation. Antioxid Redox Signal 2013; 19:1047-62. [PMID: 23311771 DOI: 10.1089/ars.2012.4535] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS Post-translational modification of proteins via carbamylation predicts increased risk for coronary artery disease. Uremia and smoke exposure are known to increase carbamylation. The aim was to investigate the role of carbamylated low-density lipoprotein (LDL) immunization on antibody formation and atherogenesis in LDL receptor-deficient (LDLR-/-) mice, and to study autoantibodies to carbamylated proteins in humans with carbamylative load. RESULTS LDLR-/- mice immunized with carbamylated mouse LDL (msLDL; n=10) without adjuvant showed specific immunoglobulin G (IgG) antibody levels to carbamyl-LDL and malondialdehyde-modified LDL (MDA-LDL) but not to oxidized LDL or native LDL. Immunization did not influence the atherosclerotic plaque area compared with control LDLR-/- mice immunized with native msLDL (n=10) or phosphate-buffered saline (n=11). Humans with high plasma urea levels, as well as smokers, had increased IgG autoantibody levels to carbamyl-modified proteins compared to the subjects with normal plasma urea levels, or to nonsmokers. INNOVATION Carbamyl-LDL induced specific IgG antibody response cross-reactive with MDA-LDL in mice. IgG antibodies to carbamyl-LDL were also found in human plasma and related to conditions known to have increased carbamylation, such as uremia and smoking. Plasma antibodies to carbamylated proteins may serve as new indicator of in vivo carbamylation. CONCLUSION These data give insight into mechanisms of in vivo humoral recognition of post-translationally modified structures. Humoral IgG immune response to carbamylated proteins is suggested to play a role in conditions leading to enhanced carbamylation, such as uremia and smoking.
Collapse
Affiliation(s)
- Outi Kummu
- 1 Department of Medical Microbiology & Immunology, Institute of Diagnostics, University of Oulu , Oulu, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Legein B, Temmerman L, Biessen EAL, Lutgens E. Inflammation and immune system interactions in atherosclerosis. Cell Mol Life Sci 2013; 70:3847-69. [PMID: 23430000 PMCID: PMC11113412 DOI: 10.1007/s00018-013-1289-1] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 16.7 million deaths each year. The underlying cause of the majority of CVD is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. Today's picture is far more complex. Atherosclerosis is considered a chronic inflammatory disease that results in the formation of plaques in large and mid-sized arteries. Both cells of the innate and the adaptive immune system play a crucial role in its pathogenesis. By transforming immune cells into pro- and anti-inflammatory chemokine- and cytokine-producing units, and by guiding the interactions between the different immune cells, the immune system decisively influences the propensity of a given plaque to rupture and cause clinical symptoms like myocardial infarction and stroke. In this review, we give an overview on the newest insights in the role of different immune cells and subtypes in atherosclerosis.
Collapse
Affiliation(s)
- Bart Legein
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lieve Temmerman
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Erik A. L. Biessen
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, Pettenkoferstrasse 8a/9, 80336 Munich, Germany
| |
Collapse
|
265
|
Krychtiuk KA, Kastl SP, Speidl WS, Wojta J. Inflammation and coagulation in atherosclerosis. Hamostaseologie 2013; 33:269-82. [PMID: 24043155 DOI: 10.5482/hamo-13-07-0039] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/09/2013] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases remain to be the leading cause of death in Western societies. Despite major findings in vascular biology that lead to a better understanding of the pathomechanisms involved in atherosclerosis, treatment of the disease has only changed slightly within the last years. A big body of evidence suggests that atherosclerosis is a chronic inflammatory disease of the vessel wall. Accumulation and peroxidation of LDL-particles within the vessel wall trigger a strong inflammatory response, causing macrophage and T-cell accumulation within the vessel wall. Additionally, B-cells and specific antibodies against LDL-particles, as well as the complement system are implicated in atherogenesis. Besides data from clinical trials and autopsy studies it was the implementation of mouse models of atherosclerosis and the emerging field of direct gen-modification that lead to a thorough description of the pathophysiological mechanisms involved in the disease and created overwhelming evidence for a participation of the immune system. Recently, the cross-talk between coagulation and inflammation in atherogenesis has gained attention. Serious limitations and disparities in the pathophysiology of atherosclerosis in mice and men complicated the translation of experimental data into clinical practice. Despite these limitations, new anti-inflammatory medical therapies in cardiovascular disease are currently being tested in clinical trials.
Collapse
Affiliation(s)
- K A Krychtiuk
- Walter S. Speidl, MD Universitätsklinik für Innere Medizin II - klinische Abteilung für Kardiologie, Medizinische Universität Wien Währingergürtel 18-20, 1090 Wien, Austria, Tel. +43/1/404 00 46 14; Fax +43/1/404 00 42 16, E-mail:
| | | | | | | |
Collapse
|
266
|
Abstract
Atherosclerosis is the primary cause of acute myocardial infarction and stroke. It is well established that arterial inflammation in response to accumulation and oxidation of lipoproteins in the vascular wall is the major factor responsible for the development of atherosclerosis. During recent years, it has become apparent that this vascular inflammation is modulated by a complex array of autoimmune responses against modified self-antigens in the atherosclerotic plaque and that both protective and pathogenic immune responses become activated as part of the disease process. Studies of hypercholesterolemia-induced immune activation in mouse models of atherosclerosis have demonstrated that Th1 cells contribute to disease progression while regulatory T cells are protective. It has been suggested that antigen presentation of modified self-antigens in the inflammatory environment of atherosclerotic plaques favors generation of antigen-specific Th1 cells over that of regulatory T cells, resulting in a local loss of tolerance. This concept has stimulated the development of plaque-antigen tolerogenic vaccines to dampen plaque inflammation and disease progression. A first generation of atherosclerosis vaccines based on peptides derived from apoB100 and heat shock proteins have demonstrated promising results in animal studies and are now approaching clinical testing.
Collapse
Affiliation(s)
- Jan Nilsson
- Department of Clinical Sciences, Malmö University Hospital, Lund University, SE-205 02 Malmö, Sweden.
| | | | | |
Collapse
|
267
|
Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guérin C, Vilar J, Caligiuri G, Tsiantoulas D, Laurans L, Dumeau E, Kotti S, Bruneval P, Charo IF, Binder CJ, Danchin N, Tedgui A, Tedder TF, Silvestre JS, Mallat Z. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med 2013; 19:1273-80. [PMID: 24037091 DOI: 10.1038/nm.3284] [Citation(s) in RCA: 436] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/26/2013] [Indexed: 12/13/2022]
Abstract
Acute myocardial infarction is a severe ischemic disease responsible for heart failure and sudden death. Here, we show that after acute myocardial infarction in mice, mature B lymphocytes selectively produce Ccl7 and induce Ly6C(hi) monocyte mobilization and recruitment to the heart, leading to enhanced tissue injury and deterioration of myocardial function. Genetic (Baff receptor deficiency) or antibody-mediated (CD20- or Baff-specific antibody) depletion of mature B lymphocytes impeded Ccl7 production and monocyte mobilization, limited myocardial injury and improved heart function. These effects were recapitulated in mice with B cell-selective Ccl7 deficiency. We also show that high circulating concentrations of CCL7 and BAFF in patients with acute myocardial infarction predict increased risk of death or recurrent myocardial infarction. This work identifies a crucial interaction between mature B lymphocytes and monocytes after acute myocardial ischemia and identifies new therapeutic targets for acute myocardial infarction.
Collapse
Affiliation(s)
- Yasmine Zouggari
- 1] Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France. [2] Université Paris-Descartes, Paris, France. [3]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
268
|
Libby P, Lichtman AH, Hansson GK. Immune effector mechanisms implicated in atherosclerosis: from mice to humans. Immunity 2013; 38:1092-104. [PMID: 23809160 DOI: 10.1016/j.immuni.2013.06.009] [Citation(s) in RCA: 517] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/12/2013] [Indexed: 02/06/2023]
Abstract
According to the traditional view, atherosclerosis results from a passive buildup of cholesterol in the artery wall. Yet, burgeoning evidence implicates inflammation and immune effector mechanisms in the pathogenesis of this disease. Both innate and adaptive immunity operate during atherogenesis and link many traditional risk factors to altered arterial functions. Inflammatory pathways have become targets in the quest for novel preventive and therapeutic strategies against cardiovascular disease, a growing contributor to morbidity and mortality worldwide. Here we review current experimental and clinical knowledge of the pathogenesis of atherosclerosis through an immunological lens and how host defense mechanisms essential for survival of the species actually contribute to this chronic disease but also present new opportunities for its mitigation.
Collapse
Affiliation(s)
- Peter Libby
- Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB7, Boston, MA 02115, USA.
| | | | | |
Collapse
|
269
|
Foks AC, Ran IA, Wasserman L, Frodermann V, Ter Borg MND, de Jager SCA, van Santbrink PJ, Yagita H, Akiba H, Bot I, Kuiper J, van Puijvelde GHM. T-cell immunoglobulin and mucin domain 3 acts as a negative regulator of atherosclerosis. Arterioscler Thromb Vasc Biol 2013; 33:2558-65. [PMID: 23990206 DOI: 10.1161/atvbaha.113.301879] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Atherosclerosis is a chronic autoimmune-like disease in which lipids and fibrous elements accumulate in the arterial blood vessels. T cells are present within atherosclerotic plaques, and their activation is partially dependent on costimulatory signals, which can either provide positive or negative signals that promote T-cell activation or limit T-cell responses, respectively. T-cell immunoglobulin and mucin domain 3 (Tim-3) is a coinhibitory type 1 transmembrane protein that affects the function of several immune cells involved in atherosclerosis, such as monocytes, macrophages, effector T cells, and regulatory T cells. In the present study, we determined the role of Tim-3 in the development of atherosclerosis. APPROACH AND RESULTS Western-type diet-fed low-density lipoprotein receptor-deficient (LDLr(-/-)) mice were treated with an anti-Tim-3 antibody for 3 and 8 weeks. Anti-Tim-3 administration increased fatty streak formation with 66% and increased atherosclerotic plaque formation after 8 weeks with 35% in the aortic root and with 50% in the aortic arch. Furthermore, blockade of Tim-3 signaling increased percentages of circulating monocytes with 33% and lesional macrophages with 20%. In addition, anti-Tim-3 administration increased CD4(+) T cells with 17%, enhanced their activation status, and reduced percentages of regulatory T cells with 18% and regulatory B cells with 37%. CONCLUSIONS It is known that Tim-3 acts as a negative regulator of both innate and adaptive immune responses, and in the present study, we show that anti-Tim-3 treatment augments lesion development, accompanied by an increase in the number of monocytes/macrophages and CD4(+) T cells and by decreased regulatory T cells and regulatory B cells.
Collapse
Affiliation(s)
- Amanda C Foks
- From the Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.C.F., I.A.R., L.W., V.F., M.N.D.t.B., S.C.A.d.J., P.J.v.S., I.B., J.K., G.H.M.v.P.); Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands (S.C.A.d.J.); and Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan (H.Y., H.A.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
270
|
Hamze M, Desmetz C, Berthe ML, Roger P, Boulle N, Brancherau P, Picard E, Guzman C, Tolza C, Guglielmi P. Characterization of Resident B Cells of Vascular Walls in Human Atherosclerotic Patients. THE JOURNAL OF IMMUNOLOGY 2013; 191:3006-16. [DOI: 10.4049/jimmunol.1202870] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
271
|
Witztum JL, Lichtman AH. The influence of innate and adaptive immune responses on atherosclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2013; 9:73-102. [PMID: 23937439 DOI: 10.1146/annurev-pathol-020712-163936] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both the chronic development of atherosclerotic lesions and the acute changes in lesion phenotype that lead to clinical cardiovascular events are significantly influenced by the innate and adaptive immune responses to lipoprotein deposition and oxidation in the arterial wall. The rapid pace of discovery of mechanisms of immunologic recognition, effector functions, and regulation has significantly influenced the study of atherosclerosis, and our new knowledge is beginning to affect how we treat this ubiquitous disease. In this review, we discuss recent advances in our understanding of how innate and adaptive immunity contribute to atherosclerosis, as well as therapeutic opportunities that arise from this knowledge.
Collapse
Affiliation(s)
- Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, California 92093;
| | | |
Collapse
|
272
|
Abstract
Cardiovascular disease is the leading cause of death in several countries. The underlying process is atherosclerosis, a slowly progressing chronic disorder that can lead to intravascular thrombosis. There is overwhelming evidence for the underlying importance of our immune system in atherosclerosis. Monocytes, which comprise part of the innate immune system, can be recruited to inflamed endothelium and this recruitment has been shown to be proportional to the extent of atherosclerotic disease. Monocytes undergo migration into the vasculature, they differentiate into macrophage phenotypes, which are highly phagocytic and can scavenge modified lipids, leading to foam cell formation and development of the lipid-rich atheroma core. This increased influx leads to a highly inflammatory environment and along with other immune cells can increase the risk in the development of the unstable atherosclerotic plaque phenotype. The present review provides an overview and description of the immunological aspect of innate and adaptive immune cell subsets in atherosclerosis, by defining their interaction with the vascular environment, modified lipids and other cellular exchanges. There is a particular focus on monocytes and macrophages, but shorter descriptions of dendritic cells, lymphocyte populations, neutrophils, mast cells and platelets are also included.
Collapse
|
273
|
TLR accessory molecule RP105 (CD180) is involved in post-interventional vascular remodeling and soluble RP105 modulates neointima formation. PLoS One 2013; 8:e67923. [PMID: 23844130 PMCID: PMC3699456 DOI: 10.1371/journal.pone.0067923] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 05/23/2013] [Indexed: 12/12/2022] Open
Abstract
Background RP105 (CD180) is TLR4 homologue lacking the intracellular TLR4 signaling domain and acts a TLR accessory molecule and physiological inhibitor of TLR4-signaling. The role of RP105 in vascular remodeling, in particular post-interventional remodeling is unknown. Methods and Results TLR4 and RP105 are expressed on vascular smooth muscle cells (VSMC) as well as in the media of murine femoral artery segments as detected by qPCR and immunohistochemistry. Furthermore, the response to the TLR4 ligand LPS was stronger in VSMC from RP105−/− mice resulting in a higher proliferation rate. In RP105−/− mice femoral artery cuff placement resulted in an increase in neointima formation as compared to WT mice (4982±974 µm2 vs.1947±278 µm2,p = 0.0014). Local LPS application augmented neointima formation in both groups, but in RP105−/− mice this effect was more pronounced (10316±1243 µm2 vs.4208±555 µm2,p = 0.0002), suggesting a functional role for RP105. For additional functional studies, the extracellular domain of murine RP105 was expressed with or without its adaptor protein MD1 and purified. SEC-MALSanalysis showed a functional 2∶2 homodimer formation of the RP105-MD1 complex. This protein complex was able to block the TLR4 response in whole blood ex-vivo. In vivo gene transfer of plasmid vectors encoding the extracellular part of RP105 and its adaptor protein MD1 were performed to initiate a stable endogenous soluble protein production. Expression of soluble RP105-MD1 resulted in a significant reduction in neointima formation in hypercholesterolemic mice (2500±573 vs.6581±1894 µm2,p<0.05), whereas expression of the single factors RP105 or MD1 had no effect. Conclusion RP105 is a potent inhibitor of post-interventional neointima formation.
Collapse
|
274
|
Mallavia B, Oguiza A, Lopez-Franco O, Recio C, Ortiz-Muñoz G, Lazaro I, Lopez-Parra V, Egido J, Gomez-Guerrero C. Gene Deficiency in Activating Fcγ Receptors Influences the Macrophage Phenotypic Balance and Reduces Atherosclerosis in Mice. PLoS One 2013; 8:e66754. [PMID: 23805273 PMCID: PMC3689671 DOI: 10.1371/journal.pone.0066754] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 05/10/2013] [Indexed: 01/18/2023] Open
Abstract
Immunity contributes to arterial inflammation during atherosclerosis. Oxidized low-density lipoproteins induce an autoimmune response characterized by specific antibodies and immune complexes in atherosclerotic patients. We hypothesize that specific Fcγ receptors for IgG constant region participate in atherogenesis by regulating the inflammatory state of lesional macrophages. In vivo we examined the role of activating Fcγ receptors in atherosclerosis progression using bone marrow transplantation from mice deficient in γ-chain (the common signaling subunit of activating Fcγ receptors) to hyperlipidemic mice. Hematopoietic deficiency of Fcγ receptors significantly reduced atherosclerotic lesion size, which was associated with decreased number of macrophages and T lymphocytes, and increased T regulatory cell function. Lesions of Fcγ receptor deficient mice exhibited increased plaque stability, as evidenced by higher collagen and smooth muscle cell content and decreased apoptosis. These effects were independent of changes in serum lipids and antibody response to oxidized low-density lipoproteins. Activating Fcγ receptor deficiency reduced pro-inflammatory gene expression, nuclear factor-κB activity, and M1 macrophages at the lesion site, while increasing anti-inflammatory genes and M2 macrophages. The decreased inflammation in the lesions was mirrored by a reduced number of classical inflammatory monocytes in blood. In vitro, lack of activating Fcγ receptors attenuated foam cell formation, oxidative stress and pro-inflammatory gene expression, and increased M2-associated genes in murine macrophages. Our study demonstrates that activating Fcγ receptors influence the macrophage phenotypic balance in the artery wall of atherosclerotic mice and suggests that modulation of Fcγ receptor-mediated inflammatory responses could effectively suppress atherosclerosis.
Collapse
Affiliation(s)
- Beñat Mallavia
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Ainhoa Oguiza
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Oscar Lopez-Franco
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Carlota Recio
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Guadalupe Ortiz-Muñoz
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Iolanda Lazaro
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Virginia Lopez-Parra
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Jesus Egido
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
| | - Carmen Gomez-Guerrero
- Renal and Vascular Inflammation Lab, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- * E-mail:
| |
Collapse
|
275
|
Affiliation(s)
- Hiroshi Iwata
- From the Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts (H.I.); Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo, Tokyo, Japan (H.I., I.M., R.N.); and Jichi Medical University, Yakushiji, Shimotsuke-shi, Tochigi Prefecture, Japan (R.N.)
| | - Ichiro Manabe
- From the Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts (H.I.); Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo, Tokyo, Japan (H.I., I.M., R.N.); and Jichi Medical University, Yakushiji, Shimotsuke-shi, Tochigi Prefecture, Japan (R.N.)
| | - Ryozo Nagai
- From the Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts (H.I.); Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo, Tokyo, Japan (H.I., I.M., R.N.); and Jichi Medical University, Yakushiji, Shimotsuke-shi, Tochigi Prefecture, Japan (R.N.)
| |
Collapse
|
276
|
Huan T, Zhang B, Wang Z, Joehanes R, Zhu J, Johnson AD, Ying S, Munson PJ, Raghavachari N, Wang R, Liu P, Courchesne P, Hwang SJ, Assimes TL, McPherson R, Samani NJ, Schunkert H, Coronary ARteryDIsease Genome wide Replication and Meta-analysis (CARDIoGRAM) Consortium International Consortium for Blood Pressure GWAS (ICBP), Meng Q, Suver C, O'Donnell CJ, Derry J, Yang X, Levy D. A systems biology framework identifies molecular underpinnings of coronary heart disease. Arterioscler Thromb Vasc Biol 2013; 33:1427-34. [PMID: 23539213 PMCID: PMC3752786 DOI: 10.1161/atvbaha.112.300112] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 03/04/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Genetic approaches have identified numerous loci associated with coronary heart disease (CHD). The molecular mechanisms underlying CHD gene-disease associations, however, remain unclear. We hypothesized that genetic variants with both strong and subtle effects drive gene subnetworks that in turn affect CHD. APPROACH AND RESULTS We surveyed CHD-associated molecular interactions by constructing coexpression networks using whole blood gene expression profiles from 188 CHD cases and 188 age- and sex-matched controls. Twenty-four coexpression modules were identified, including 1 case-specific and 1 control-specific differential module (DM). The DMs were enriched for genes involved in B-cell activation, immune response, and ion transport. By integrating the DMs with gene expression-associated single-nucleotide polymorphisms and with results of genome-wide association studies of CHD and its risk factors, the control-specific DM was implicated as CHD causal based on its significant enrichment for both CHD and lipid expression-associated single-nucleotide polymorphisms. This causal DM was further integrated with tissue-specific Bayesian networks and protein-protein interaction networks to identify regulatory key driver genes. Multitissue key drivers (SPIB and TNFRSF13C) and tissue-specific key drivers (eg, EBF1) were identified. CONCLUSIONS Our network-driven integrative analysis not only identified CHD-related genes, but also defined network structure that sheds light on the molecular interactions of genes associated with CHD risk.
Collapse
Affiliation(s)
- Tianxiao Huan
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biological Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Zhi Wang
- Sage Bionetworks, 1100 Fairview Ave N, Seattle, WA 98109, USA
- School of Life Sciences, Center for Evolutionary Medicine and Informatics, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Roby Joehanes
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
- Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biological Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew D. Johnson
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Saixia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, USA
| | - Peter J. Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, USA
| | | | - Richard Wang
- Genomics Core facility Genetics & Developmental Biology Center, NHLBI, USA
| | - Poching Liu
- Genomics Core facility Genetics & Developmental Biology Center, NHLBI, USA
| | - Paul Courchesne
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Shih-Jen Hwang
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - Ruth McPherson
- Departments of Medicine and Biochemistry, University of Ottawa, USA
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Leicester, UK
| | - Heribert Schunkert
- Medizinische Klinik II, Universitätzu Lübeck, Lübeck, and Deutsches Zentrumfür Herz-Kreislauf-Forschung (DZHK), Universitätzu Lübeck, Lübeck, Germany
| | | | - Qingying Meng
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christine Suver
- Sage Bionetworks, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Christopher J. O'Donnell
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jonathan Derry
- Sage Bionetworks, 1100 Fairview Ave N, Seattle, WA 98109, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Levy
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA 01702, USA
- The Center for Population Studies and the Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| |
Collapse
|
277
|
Sternberg Z, Ghanim H, Gillotti KM, Tario JD, Munschauer F, Curl R, Noor S, Yu J, Ambrus JL, Wallace P, Dandona P. Flow cytometry and gene expression profiling of immune cells of the carotid plaque and peripheral blood. Atherosclerosis 2013; 229:338-47. [PMID: 23880185 DOI: 10.1016/j.atherosclerosis.2013.04.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/08/2013] [Accepted: 04/26/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVES The relative contribution of the local vs. peripheral inflammation to the atherothrombotic processes is unknown. We compared the inflammatory status of the immune cells of the carotid plaque with similar cells in peripheral circulation of patients with advanced carotid disease (PCDs). METHODS Mononuclear cells (MNCs) were extracted from carotid endarterectomy (CEA) samples by enzymatic digestion and subsequent magnetic cell sorting. The cell surface antigenic expressions, and mRNA expression levels were compared between CEA MNCs and peripheral MNCs, using flow cytometry and RT-PCR techniques. RESULTS The percentages of resting MNCs were lower, and activated MNCs, particularly monocytes, were higher in the CEAMNCs, as compared to the peripheral MNCs. The percentages of activated T cells and B cells were higher in the peripheral MNCs of PCDs, than in healthy controls (HCs), but the percentages of activated monocytes did not differ between the two groups. The expression levels of both pro-inflammatory/pro-thrombotic (P(38), JNKB-1, Egr-1 PAI-1, MCP-1, TF, MMP-9, HMGB-1, TNF-α, mTOR) and anti-inflammatory (PPAR-γ, TGF-β) mediators were significantly higher in the CEA MNCs as compared to the peripheral MNCs. Furthermore, MMP-9 and PPAR-γ expression levels were higher in the peripheral MNCs of PCDs than HCs. CONCLUSION The inflammatory status is higher in the immune cells of the carotid plaque, as compared to those cells in the peripheral blood. The altered expression levels of both pro-inflammatory/pro-thrombotic and anti-inflammatory mediators in the milieu of the plaque suggest that the balance between these various mediators may play a key role in carotid disease progression.
Collapse
|
278
|
Obesity Modulates the Immune Response to Oxidized LDL in Hypertensive Patients. Cell Biochem Biophys 2013; 67:1451-60. [DOI: 10.1007/s12013-013-9585-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
279
|
Kyaw T, Cui P, Tay C, Kanellakis P, Hosseini H, Liu E, Rolink AG, Tipping P, Bobik A, Toh BH. BAFF receptor mAb treatment ameliorates development and progression of atherosclerosis in hyperlipidemic ApoE(-/-) mice. PLoS One 2013; 8:e60430. [PMID: 23560095 PMCID: PMC3616162 DOI: 10.1371/journal.pone.0060430] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 02/25/2013] [Indexed: 01/19/2023] Open
Abstract
Aims Option to attenuate atherosclerosis by depleting B2 cells is currently limited to anti-CD20 antibodies which deplete all B-cell subtypes. In the present study we evaluated the capacity of a monoclonal antibody to B cell activating factor-receptor (BAFFR) to selectively deplete atherogenic B2 cells to prevent both development and progression of atherosclerosis in the ApoE−/− mouse. Methods and Results To determine whether the BAFFR antibody prevents atherosclerosis development, we treated ApoE−/− mice with the antibody while feeding them a high fat diet (HFD) for 8 weeks. Mature CD93− CD19+ B2 cells were reduced by treatment, spleen B-cell zones disrupted and spleen CD20 mRNA expression decreased while B1a cells and non-B cells were spared. Atherosclerosis was ameliorated in the hyperlipidemic mice and CD19+ B cells, CD4+ and CD8+ T cells were reduced in atherosclerotic lesions. Expressions of proinflammatory cytokines, IL1β, TNFα, and IFNγ in the lesions were also reduced, while MCP1, MIF and VCAM-1 expressions were unaffected. Plasma immunoglobulins were reduced, but MDA-oxLDL specific antibodies were unaffected. To determine whether anti-BAFFR antibody ameliorates progression of atherosclerosis, we first fed ApoE−/− mice a HFD for 6 weeks, and then instigated anti-BAFFR antibody treatment for a further 6 week-HFD. CD93− CD19+ B2 cells were selectively decreased and atherosclerotic lesions were reduced by this treatment. Conclusion Anti-BAFFR monoclonal antibody selectively depletes mature B2 cells while sparing B1a cells, disrupts spleen B-cell zones and ameliorates atherosclerosis development and progression in hyperlipidemic ApoE−/− mice. Our findings have potential for clinical translation to manage atherosclerosis-based cardiovascular diseases.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory Baker IDI Heart and Diabetes Institute, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
280
|
Rituximab and vascular function in granulomatosis with polyangiitis (GPA). Presse Med 2013. [DOI: 10.1016/j.lpm.2013.02.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
281
|
Benucci M, Saviola G, Manfredi M, Sarzi-Puttini P, Atzeni F. Factors correlated with improvement of endothelial dysfunction during rituximab therapy in patients with rheumatoid arthritis. Biologics 2013; 7:69-75. [PMID: 23526116 PMCID: PMC3603333 DOI: 10.2147/btt.s39182] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Increased cardiovascular mortality has been associated with rheumatoid arthritis (RA). There are reports indicating that tumor necrosis factor (TNF) blockers may exert favorable but transient effects on the lipid profile, flow-mediated vasodilatation (FMD) of the brachial artery, and the common carotid intima–media thickness (ccIMT) in RA. We evaluated 38 RA patients (33 females and five males with a mean age of 66.7 ± 10.2 years) who were unresponsive to TNF blockers. The patients received one or more courses of two rituximab (RTX) 1000 mg infusions. Disease activity was evaluated at each visit. Investigations included erythrocyte sedimentation rate, C-reactive protein (CRP) levels, the 28-joint disease activity score (DAS28), DAS28CRP, the Health Assessment Questionnaire, the FMD percent change from baseline (FMD%), and the postnitroglycerine endothelium-independent vasodilatation. In comparison with the baseline, there was a significant improvement in clinical variables and acute-phase reactants 24 months after the start of RTX therapy. There was also a major improvement in FMD% (from baseline 5.24 ± 1.12 to 5.43 ± 1.16; P = −0.03) and a smaller change in the ccIMT (from baseline 0.69 ± 0.16 to 0.67 ± 0.12 mm P = 0.25). Univariate analysis showed that global health (P < 0.034) was associated with the improvement in FMD%. Multivariate models showed that GH (odds ratio [OR] 0.91; 95% CI: 0.99–0.83; P = 0.032), CD19+ cells (OR 1.024; 95% CI: 1.045–1.003; P = 0.025), IgM (OR 1.025; 95% CI: 1.045–1.004; P = 0.016), and interleukin (IL)-8 (OR 0.487; 95% CI: 0.899–0.264; P = 0.021) were statistically associated with the improvement of FMD%, and that IL-8 (OR 0.717; 95% CI: 0.926–0.555; P = 0.018) was also statistically associated with improvement of ccIMT. The findings of the study confirm that RTX reduces the progression of accelerated atherosclerosis in patients with RA. They also show that improvement in CD19+ cells, IgM and GH after treatment are statistically associated with the improvement of FMD%, and that improvement in IL-8 levels after treatment is statistically associated with improved FMD% and with decrease in the ccIMT.
Collapse
Affiliation(s)
- Maurizio Benucci
- Rheumatology Unit, Department of Internal Medicine, Hospital di S Giovanni di Dio, Azienda Sanitaria di Firenze, Florence, Italy
| | | | | | | | | |
Collapse
|
282
|
Nikolajczyk BS, Jagannathan-Bogdan M, Denis GV. The outliers become a stampede as immunometabolism reaches a tipping point. Immunol Rev 2013; 249:253-75. [PMID: 22889227 DOI: 10.1111/j.1600-065x.2012.01142.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity and Type 2 diabetes mellitus (T2D) are characterized by pro-inflammatory alterations in the immune system including shifts in leukocyte subset differentiation and in cytokine/chemokine balance. The chronic, low-grade inflammation resulting largely from changes in T-cell, B-cell, and myeloid compartments promotes and/or exacerbates insulin resistance (IR) that, together with pancreatic islet failure, defines T2D. Animal model studies show that interruption of immune cell-mediated inflammation by any one of several methods almost invariably results in the prevention or delay of obesity and/or IR. However, anti-inflammatory therapies have had a modest impact on established T2D in clinical trials. These seemingly contradictory results indicate that a more comprehensive understanding of human IR/T2D-associated immune cell function is needed to leverage animal studies into clinical treatments. Important outstanding analyses include identifying potential immunological checkpoints in disease etiology, detailing immune cell/adipose tissue cross-talk, and defining strengths/weaknesses of model organism studies to determine whether we can harness the promising new field of immunometabolism to curb the global obesity and T2D epidemics.
Collapse
|
283
|
Lefaucheur C, Loupy A, Vernerey D, Duong-Van-Huyen JP, Suberbielle C, Anglicheau D, Vérine J, Beuscart T, Nochy D, Bruneval P, Charron D, Delahousse M, Empana JP, Hill GS, Glotz D, Legendre C, Jouven X. Antibody-mediated vascular rejection of kidney allografts: a population-based study. Lancet 2013. [PMID: 23182298 DOI: 10.1016/s0140-6736(12)61265-3] [Citation(s) in RCA: 264] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Rejection of allografts has always been the major obstacle to transplantation success. We aimed to improve characterisation of different kidney-allograft rejection phenotypes, identify how each one is associated with anti-HLA antibodies, and investigate their distinct prognoses. METHODS Patients who underwent ABO-compatible kidney transplantations in Necker Hospital and Saint-Louis Hospital (Paris, France) between Jan 1, 1998, and Dec 31, 2008, were included in our population-based study. We assessed patients who provided biopsy samples for acute allograft rejection, which was defined as the association of deterioration in function and histopathological lesions. The main outcome was kidney allograft loss-ie, return to dialysis. To investigate distinct rejection patterns, we retrospectively assessed rejection episodes with review of graft histology, C4d in allograft biopsies, and donor-specific anti-HLA antibodies. FINDINGS 2079 patients were included in the main analyses, of whom 302 (15%) had acute biopsy-proven rejection. We identified four distinct patterns of kidney allograft rejection: T cell-mediated vascular rejection (26 patients [9%]), antibody-mediated vascular rejection (64 [21%]), T cell-mediated rejection without vasculitis (139 [46%]), and antibody-mediated rejection without vasculitis (73 [24%]). Risk of graft loss was 9·07 times (95 CI 3·62-19·7) higher in antibody-mediated vascular rejection than in T cell-mediated rejection without vasculitis (p<0·0001), compared with an increase of 2·93 times (1·1-7·9; P=0·0237) in antibody-mediated rejection without vasculitis and no significant rise in T cell-mediated vascular rejection (hazard ratio [HR] 1·5, 95% CI 0·33-7·6; p=0·60). INTERPRETATION We have identified a type of kidney rejection not presently included in classifications: antibody-mediated vascular rejection. Recognition of this distinct phenotype could lead to the development of new treatment strategies that could salvage many kidney allografts. FUNDING None.
Collapse
Affiliation(s)
- Carmen Lefaucheur
- Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique Hôpitaux de Paris, INSERM U940, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Miller VM, Petterson TM, Jeavons EN, Lnu AS, Rider DN, Heit JA, Cunningham JM, Huggins GS, Hodis HN, Budoff MJ, Santoro N, Hopkins PN, Lobo RA, Manson JE, Naftolin F, Taylor HS, Harman SM, de Andrade M. Genetic polymorphisms associated with carotid artery intima-media thickness and coronary artery calcification in women of the Kronos Early Estrogen Prevention Study. Physiol Genomics 2013; 45:79-88. [PMID: 23188791 PMCID: PMC3546410 DOI: 10.1152/physiolgenomics.00114.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 11/25/2012] [Indexed: 12/15/2022] Open
Abstract
Menopausal hormone treatment (MHT) may limit progression of cardiovascular disease (CVD) but poses a thrombosis risk. To test targeted candidate gene variation for association with subclinical CVD defined by carotid artery intima-media thickness (CIMT) and coronary artery calcification (CAC), 610 women participating in the Kronos Early Estrogen Prevention Study (KEEPS), a clinical trial of MHT to prevent progression of CVD, were genotyped for 13,229 single nucleotide polymorphisms (SNPs) within 764 genes from anticoagulant, procoagulant, fibrinolytic, or innate immunity pathways. According to linear regression, proportion of European ancestry correlated negatively, but age at enrollment and pulse pressure correlated positively with CIMT. Adjusting for these variables, two SNPs, one on chromosome 2 for MAP4K4 gene (rs2236935, β = 0.037, P value = 2.36 × 10(-06)) and one on chromosome 5 for IL5 gene (rs739318, β = 0.051, P value = 5.02 × 10(-05)), associated positively with CIMT; two SNPs on chromosome 17 for CCL5 (rs4796119, β = -0.043, P value = 3.59 × 10(-05); rs2291299, β = -0.032, P value = 5.59 × 10(-05)) correlated negatively with CIMT; only rs2236935 remained significant after correcting for multiple testing. Using logistic regression, when we adjusted for waist circumference, two SNPs (rs11465886, IRAK2, chromosome 3, OR = 3.91, P value = 1.10 × 10(-04); and rs17751769, SERPINA1, chromosome 14, OR = 1.96, P value = 2.42 × 10(-04)) associated positively with a CAC score of >0 Agatston unit; one SNP (rs630014, ABO, OR = 0.51, P value = 2.51 × 10(-04)) associated negatively; none remained significant after correcting for multiple testing. Whether these SNPs associate with CIMT and CAC in women randomized to MHT remains to be determined.
Collapse
|
285
|
Lichtman AH, Binder CJ, Tsimikas S, Witztum JL. Adaptive immunity in atherogenesis: new insights and therapeutic approaches. J Clin Invest 2013; 123:27-36. [PMID: 23281407 DOI: 10.1172/jci63108] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many remarkable advances have improved our understanding of the cellular and molecular events in the pathogenesis of atherosclerosis. Chief among these is the accumulating knowledge of how the immune system contributes to all phases of atherogenesis, including well-known inflammatory reactions consequent to intimal trapping and oxidation of LDL. Advances in our understanding of the innate and adaptive responses to these events have helped to clarify the role of inflammation in atherogenesis and suggested new diagnostic modalities and novel therapeutic targets. Here we focus on recent advances in understanding how adaptive immunity affects atherogenesis.
Collapse
Affiliation(s)
- Andrew H Lichtman
- Vascular Research Division, Department of Pathology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
286
|
Pastrana JL, Sha X, Virtue A, Mai J, Cueto R, Lee IA, Wang H, Yang XF. Regulatory T cells and Atherosclerosis. ACTA ACUST UNITED AC 2013; 2012:2. [PMID: 23997979 DOI: 10.4172/2155-9880.s12-002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a chronic autoimmune inflammatory disease. The involvement of both innate and adaptive immune responses in the pathogenesis of the disease has been well recognized. Tregs are an essential part of the immune system and have indispensable functions in maintaining immune system homeostasis, mediating peripheral tolerance, preventing autoimmune diseases, and suppressing inflammatory and proatherogenic immune response. Tregs carry out their immunosuppressive functions via several mechansims. One of the well-documented suppressive mechanisms of Tregs is the secretion of anti-inflammatory cytokines including IL-10, TGF-β, and IL-35. Studies have found that IL-10 and TGF-β have atheroprotective properties. In addition, Tregs can suppress the activity of proatherogenic effector T cells, suggesting an atheroprotective role. In fact, fewer Tregs are found in atherogenic ApoE-/- mice comparing to wild-type mice, suggesting an uncontrolled balance between weakened Tregs and effector T cells in atherogenesis. Some clinical studies of autoimmune diseases also suggest that decreased Tregs numbers are associated with increased disease activity. The importance of Tregs in many autoimmune diseases and experimental atherosclerosis has been established in in vivo and in vitro studies. However, the roles of Tregs in atherosclerosis in the clinical setting remains to be further characterized.
Collapse
Affiliation(s)
- Jahaira Lopez Pastrana
- Cardiovascular Research Center, Department of Pharmacology and Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140
| | | | | | | | | | | | | | | |
Collapse
|
287
|
Perry HM, Bender TP, McNamara CA. B cell subsets in atherosclerosis. Front Immunol 2012; 3:373. [PMID: 23248624 PMCID: PMC3518786 DOI: 10.3389/fimmu.2012.00373] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/21/2012] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis, the underlying cause of heart attacks and strokes, is a chronic inflammatory disease of the artery wall. Immune cells, including lymphocytes modulate atherosclerotic lesion development through interconnected mechanisms. Elegant studies over the past decades have begun to unravel a role for B cells in atherosclerosis. Recent findings provide evidence that B cell effects on atherosclerosis may be subset-dependent. B-1a B cells have been reported to protect from atherosclerosis by secretion of natural IgM antibodies. Conventional B-2 B cells can promote atherosclerosis through less clearly defined mechanism that may involve CD4 T cells. Yet, there may be other populations of B cells within these subsets with different phenotypes altering their impact on atherosclerosis. Additionally, the role of B cell subsets in atherosclerosis may depend on their environmental niche and/or the stage of atherogenesis. This review will highlight key findings in the evolving field of B cells and atherosclerosis and touch on the potential and importance of translating these findings to human disease.
Collapse
Affiliation(s)
- Heather M Perry
- Department of Pathology, University of Virginia Charlottesville, VA, USA ; Cardiovascular Research Center, University of Virginia Health System Charlottesville, VA, USA
| | | | | |
Collapse
|
288
|
Increased inflammation in atherosclerotic lesions of diabetic Akita-LDLr⁻/⁻ mice compared to nondiabetic LDLr⁻/⁻ mice. EXPERIMENTAL DIABETES RESEARCH 2012; 2012:176162. [PMID: 23243415 PMCID: PMC3515907 DOI: 10.1155/2012/176162] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/09/2012] [Accepted: 10/12/2012] [Indexed: 12/19/2022]
Abstract
Background. Diabetes is associated with increased cardiovascular disease, but the underlying cellular and molecular mechanisms are poorly understood. One proposed mechanism is that diabetes aggravates atherosclerosis by enhancing plaque inflammation. The Akita mouse has recently been adopted as a relevant model for microvascular complications of diabetes. Here we investigate the development of atherosclerosis and inflammation in vessels of Akita mice on LDLr−/− background. Methods and Results. Akita-LDLr−/− and LDLr−/− mice were fed high-fat diet from 6 to 24 weeks of age. Blood glucose levels were higher in both male and female Akita-LDLr−/− mice (137% and 70%, resp.). Male Akita-LDLr−/− mice had markedly increased plasma cholesterol and triglyceride levels, a three-fold increase in atherosclerosis, and enhanced accumulation of macrophages and T-cells in plaques. In contrast, female Akita-LDLr−/− mice demonstrated a modest 29% increase in plasma cholesterol and no significant increase in triglycerides, atherosclerosis, or inflammatory cells in lesions. Male Akita-LDLr−/− mice had increased levels of plasma IL-1β compared to nondiabetic mice, whereas no such difference was seen between female diabetic and nondiabetic mice. Conclusion. Akita-LDLr−/− mice display considerable gender differences in the development of diabetic atherosclerosis. In addition, the increased atherosclerosis in male Akita-LDLr−/− mice is associated with an increase in inflammatory cells in lesions.
Collapse
|
289
|
Businaro R. Neuroimmunology of the atherosclerotic plaque: a morphological approach. J Neuroimmune Pharmacol 2012; 8:15-27. [PMID: 23150034 DOI: 10.1007/s11481-012-9421-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 11/05/2012] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is a chronic inflammatory process, lasting for several decades until the onset of its clinical manifestations. The progression of the atherosclerotic lesion to a stable fibrotic plaque, narrowing the vascular lumen, or to a vulnerable plaque leading to main vascular complications, is associated to the involvement of several cell subpopulations of the innate as well as of the adaptive immunity, and to the release of chemokines and pro-inflammatory cytokines. Emerging evidence outlines that the cardiovascular risk is dependent on stress-mediators influencing cell migration and vascular remodeling. The view that atherosclerosis is initiated by monocytes and lymphocytes adhering to dysfunctional endothelial cells is substantiated by experimental and clinical observations. Macrophages, dendritic cells, T and B lymphocytes, granulocytes accumulating into the subendothelial space secrete and are stimulated by soluble factors, including peptides, proteases and cytokines acting synergistically. The final step of the disease, leading to plaque destabilization and rupture, is induced by the release, at the level of the fibrous cap, of metalloproteinases and elastases by the activated leukocytes which accumulate locally. Recruitment of specific cell subpopulations as well as the progression of atherosclerotic lesions towards a stable or an unstable phenotype, are related to the unbalance between pro-atherogenic and anti-atherogenic factors. In this connection stress hormones deserve particular attention, since their role in vascular remodeling, via vascular smooth cell proliferation, as well as in neoangiogenesis, via stimulation of endothelial cell proliferation and migration, has been already established.
Collapse
Affiliation(s)
- Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy.
| |
Collapse
|
290
|
Francosalinas G, Cantaert T, Nolte MA, Tak PP, van Lier RAW, Baeten DL. Enhanced costimulation by CD70+ B cells aggravates experimental autoimmune encephalomyelitis in autoimmune mice. J Neuroimmunol 2012; 255:8-17. [PMID: 23137837 DOI: 10.1016/j.jneuroim.2012.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 10/12/2012] [Accepted: 10/16/2012] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Assess whether CD70+ B cells contribute to EAE. MATERIALS AND METHODS MOG-specific TCR transgenic mice (2D2) were crossed with mice with constitutive CD70 expression on B cells. The development of EAE and the phenotype of B-T lymphocytes were studied in 2D2xCD70 animals. RESULTS Spontaneous EAE developed in 20% of 2D2xCD70 and 3% of 2D2 mice. EAE was also more severe in 2D2xCD70 versus 2D2 animals upon MOG immunization. The susceptibility of 2D2xCD70 to EAE was associated with fewer FoxP3+ T cells. CONCLUSIONS Expression of CD70 by B cells aggravates EAE possibly by reducing the number of regulatory T cells.
Collapse
Affiliation(s)
- G Francosalinas
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
291
|
Emerging biomarkers and intervention targets for immune-modulation of atherosclerosis - a review of the experimental evidence. Atherosclerosis 2012. [PMID: 23177975 DOI: 10.1016/j.atherosclerosis.2012.10.074] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The role of inflammation in atherosclerosis and plaque vulnerability is well recognized. However, it is only during recent years it has become evident that this inflammation is modulated by immune responses against plaque antigens such as oxidized LDL. Interestingly, both protective and pathogenic immune responses exist and experimental data from animal studies suggest that modulation of these immune responses represents a promising new target for treatment of cardiovascular disease. It has been proposed that during early stages of the disease, autoimmune responses against plaque antigens are controlled by regulatory T cells that inhibit the activity of auto-reactive Th1 effector T cells by release of anti-inflammatory cytokines such as IL-10 and TGF-β. As the disease progresses this control is gradually lost and immune responses towards plaque antigens switch towards activation of Th1 effector T cells and release of pro-inflammatory cytokines such as interferon-γ, TNF-α and IL-1β. Several novel immune-modulatory therapies that promote or mimic tolerogenic immune responses against plaque antigens have demonstrated athero-protective effects in experimental models and a first generation of such immune-modulatory therapies are now in early or about to enter into clinical testing. A challenge in the clinical development of these therapies is that our knowledge of the role of the immune system in atherosclerosis largely rests on data from animal models of the disease. It is therefore critical that more attention is given to the characterization and evaluation of immune biomarkers for cardiovascular risk.
Collapse
|
292
|
Abstract
Experimental work has elucidated molecular and cellular pathways of inflammation that promote atherosclerosis. Unraveling the roles of cytokines as inflammatory messengers provided a mechanism whereby risk factors for atherosclerosis can alter arterial biology, and produce a systemic milieu that favors atherothrombotic events. The discovery of the immune basis of allograft arteriosclerosis demonstrated that inflammation per se can drive arterial hyperplasia, even in the absence of traditional risk factors. Inflammation regulates aspects of plaque biology that trigger the thrombotic complications of atherosclerosis. Translation of these discoveries to humans has enabled both novel mechanistic insights and practical clinical advances.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
293
|
Kaminski DA, Wei C, Qian Y, Rosenberg AF, Sanz I. Advances in human B cell phenotypic profiling. Front Immunol 2012; 3:302. [PMID: 23087687 PMCID: PMC3467643 DOI: 10.3389/fimmu.2012.00302] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/10/2012] [Indexed: 12/11/2022] Open
Abstract
To advance our understanding and treatment of disease, research immunologists have been called-upon to place more centralized emphasis on impactful human studies. Such endeavors will inevitably require large-scale study execution and data management regulation (“Big Biology”), necessitating standardized and reliable metrics of immune status and function. A well-known example setting this large-scale effort in-motion is identifying correlations between eventual disease outcome and T lymphocyte phenotype in large HIV-patient cohorts using multiparameter flow cytometry. However, infection, immunodeficiency, and autoimmunity are also characterized by correlative and functional contributions of B lymphocytes, which to-date have received much less attention in the human Big Biology enterprise. Here, we review progress in human B cell phenotyping, analysis, and bioinformatics tools that constitute valuable resources for the B cell research community to effectively join in this effort.
Collapse
Affiliation(s)
- Denise A Kaminski
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Rochester, NY, USA
| | | | | | | | | |
Collapse
|
294
|
Wilhelm AJ, Major AS. Accelerated atherosclerosis in SLE: mechanisms and prevention approaches. ACTA ACUST UNITED AC 2012; 7:527-539. [PMID: 24672580 DOI: 10.2217/ijr.12.46] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by increased serum autoantibody levels and tissue damage. With improved diagnosis and more effective treatment of the resultant kidney disease, accelerated atherosclerosis has become a major cause of morbidity in patients suffering from SLE. Although the exact mechanisms for SLE-accelerated atherosclerosis are unknown, multiple factors have been established as potential players in this process. Among these potential players are dysregulation of T and B cell populations and increased circulating levels of inflammatory cytokines. In addition, SLE patients exhibit a proatherogenic lipid profile characterized by low HDL and high LDL and triglycerides. Recent therapeutic approaches have focused on targeting B cells, the producers of autoantibodies, but most studies do not consider the effects of these treatments on atherosclerosis. Evidence suggests that T cells play a major role in SLE-accelerated atherosclerosis. Therefore, therapies targeted at T cells may also prove invaluable in treating SLE and atherosclerosis.
Collapse
Affiliation(s)
- Ashley J Wilhelm
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amy S Major
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
295
|
Grönberg C, Björkbacka H. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol 2012; 23:505-8. [PMID: 22964998 DOI: 10.1097/mol.0b013e32835821d2] [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: 11/26/2022]
|
296
|
|
297
|
Lopez-Parra V, Mallavia B, Lopez-Franco O, Ortiz-Muñoz G, Oguiza A, Recio C, Blanco J, Nimmerjahn F, Egido J, Gomez-Guerrero C. Fcγ receptor deficiency attenuates diabetic nephropathy. J Am Soc Nephrol 2012; 23:1518-27. [PMID: 22859852 DOI: 10.1681/asn.2011080822] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Among patients with diabetes, increased production of immunoglobulins against proteins modified by diabetes is associated with proteinuria and cardiovascular risk, suggesting that immune mechanisms may contribute to the development of diabetes complications, such as nephropathy. We investigated the contribution of IgG Fcγ receptors to diabetic renal injury in hyperglycemic, hypercholesterolemic mice. We used streptozotocin to induce diabetes in apolipoprotein E-deficient mice and in mice deficient in both apolipoprotein E and γ-chain, the common subunit of activating Fcγ receptors. After 15 weeks, the mice lacking Fcγ receptors had significantly less albuminuria and renal hypertrophy, despite similar degrees of hyperglycemia and hypercholesterolemia, immunoglobulin production, and glomerular immune deposits. Moreover, diabetic Fcγ receptor-deficient mice had less mesangial matrix expansion, inflammatory cell infiltration, and collagen and α-smooth muscle actin content in their kidneys. Accordingly, expression of genes involved in leukocyte infiltration, fibrosis, and oxidative stress was significantly reduced in diabetic kidneys and in mesangial cells cultured from Fcγ receptor-deficient mice. In summary, preventing the activation of Fcγ receptors alleviates renal hypertrophy, inflammation, and fibrosis in hypercholesterolemic mice with diabetes, suggesting that modulating Fcγ receptor signaling may be renoprotective in diabetic nephropathy.
Collapse
Affiliation(s)
- Virginia Lopez-Parra
- Renal and Vascular Inflammation Laboratory, IIS-Fundacion Jimenez Diaz, Autonoma University, Avda Reyes Catolicos, 2 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
298
|
Abstract
Atherosclerosis initiated by hyperlipidemia is modulated by immune cells in its development, progression, and rupture that results in thrombotic arterial occlusion leading to strokes and myocardial infarction. B cells initially thought to be atheroprotective provide opposing roles by their different subsets. Unlike B2 cells that are atherogenic, serosal B1a cells are atheroprotective by producing natural IgM antibodies that clear modified low-density lipoprotein and apoptotic and necrotic debris. In addition to natural IgM antibodies, B1a cells may act as regulatory B cells by producing the anti-inflammatory cytokine interleukin-10, which inhibits proinflammatory cytokines secreted by activated macrophages and T cells in atherosclerotic lesions. These findings suggest in vivo expansion of atheroprotective B1a cells as a potential therapeutic strategy to augment the benefits of lipid-lowering statin therapy.
Collapse
|
299
|
Abstract
Obesity is associated with chronic inflammation of various tissues including visceral adipose tissue (VAT), which contributes to insulin resistance. T cells and macrophages infiltrate VAT in obesity and orchestrate this inflammation. Recently, we made the surprising discovery that B cells are important contributors to this process. Thus, some B cells and the antibodies they produce can promote VAT-associated and systemic inflammation, leading to insulin resistance. This report will focus on the properties of these B cells, and how they contribute to insulin resistance through T-cell modulation and production of pathogenic autoantibodies. Understanding the mechanisms by which B cells contribute to insulin resistance should lead to new antibody-based diagnostics and B-cell modulating therapeutics to manage this increasingly prevalent disease.
Collapse
|
300
|
Weih F, Gräbner R, Hu D, Beer M, Habenicht AJR. Control of dichotomic innate and adaptive immune responses by artery tertiary lymphoid organs in atherosclerosis. Front Physiol 2012; 3:226. [PMID: 22783198 PMCID: PMC3390894 DOI: 10.3389/fphys.2012.00226] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 06/04/2012] [Indexed: 12/29/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) emerge in tissues in response to non-resolving inflammation such as chronic infection, graft rejection, and autoimmune disease. We identified artery TLOs (ATLOs) in the adventitia adjacent to atherosclerotic plaques of aged hyperlipidemic ApoE−/− mice. ATLOs are structured into T cell areas harboring conventional dendritic cells and monocyte-derived DCs; B cell follicles containing follicular dendritic cells within activated germinal centers; and peripheral niches of plasma cells. ATLOs also show extensive neoangiogenesis, aberrant lymphangiogenesis, and high endothelial venule (HEV) neogenesis. Newly formed conduit networks connect the external lamina of the artery with HEVs in T cell areas. ATLOs recruit and generate lymphocyte subsets with opposing activities including activated CD4+ and CD8+ effector T cells, natural and induced CD4+ T regulatory (nTregs; iTregs) cells as well as B-1 and B-2 cells at different stages of differentiation. These data indicate that ATLOs organize dichotomic innate and adaptive immune responses in atherosclerosis. In this review we discuss the novel concept that dichotomic immune responses toward atherosclerosis-specific antigens are carried out by ATLOs in the adventitia of the arterial wall and that malfunction of the tolerogenic arm of ATLO immunity triggers transition from silent autoimmune reactivity to clinically overt disease.
Collapse
Affiliation(s)
- Falk Weih
- Leibniz-Institute for Age Research, Fritz-Lipmann-Institute Jena, Germany
| | | | | | | | | |
Collapse
|