1
|
Herrock O, Campbell N, Deer E, Amaral LM, Whitney D, Morris R, Wallace K, Turner T, Cleveland EH, Belk S, Booz GW, Cornelius DC, LaMarca B. Preeclamptic Placental CD19+ B Cells Are Causal to Hypertension During Pregnancy. Hypertension 2025; 82:894-903. [PMID: 40171666 PMCID: PMC12003061 DOI: 10.1161/hypertensionaha.124.24552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Patients with preeclampsia exhibit hypertension and chronic inflammation characterized by CD (cluster determinant) 4+T cells, B cells secreting AT1-AA (agonistic autoantibody against the angiotensin II type 1 receptor), inflammatory cytokines, and complement activation. Importantly, a history of COVID-19 during pregnancy is associated with an increased incidence of a preeclampsia-like phenotype and is partly mediated by CD4+T cells. We recently showed pregnant patients with a history of COVID-19 with or without preeclampsia produce AT1-AA, indicating the importance of B lymphocytes in the progression of preeclampsia and possibly of COVID-19. Therefore, we hypothesize that B cells from patients with preeclampsia with or without COVID-19 history induce the preeclampsia phenotype through AT1-AA. METHODS Placental B cells were isolated from normal pregnant, patients with preeclampsia, normotensive COVID-19 history, or preeclampsia COVID-19 history at delivery. Then, 3×105 B cells were transferred intraperitoneally into pregnant athymic rats at gestational day 12. On gestational day 18, carotid catheters were inserted. On gestational day 19, mean arterial pressure was measured, and tissues were collected. RESULTS Preeclampsia B-cell recipients had significantly increased mean arterial pressure, AT1-AA, inflammatory cytokines, and complement activation compared with normal pregnant B-cell recipients. Recipients of B cells with COVID-19 history had markers of inflammation and hypertension but not to the level of significance as recipients of preeclampsia B cells. Inhibition of AT1-AA attenuated the hypertension that occurred in response to preeclampsia or preeclampsia B cells with COVID-19 history. CONCLUSIONS This study demonstrates the important role of B cells in contributing to hypertension and chronic inflammation during preeclampsia with or without COVID-19 history through secretion of AT1-AA.
Collapse
Affiliation(s)
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Lorena M. Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Darby Whitney
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Rachael Morris
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - E. Hawthorne Cleveland
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Sheila Belk
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - George W. Booz
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Denise C. Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
2
|
Goh WX, Kok YY, Wong CY. Comparison of Cell-based and Nanoparticle-based Therapeutics in Treating Atherosclerosis. Curr Pharm Des 2023; 29:2827-2840. [PMID: 37936453 DOI: 10.2174/0113816128272185231024115046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 11/09/2023]
Abstract
Today, cardiovascular diseases are among the biggest public health threats worldwide. Atherosclerosis, a chronic inflammatory disease with complex aetiology and pathogenesis, predispose many of these conditions, including the high mortality rate-causing ischaemic heart disease and stroke. Nevertheless, despite the alarming prevalence and absolute death rate, established treatments for atherosclerosis are unsatisfactory in terms of efficacy, safety, and patient acceptance. The rapid advancement of technologies in healthcare research has paved new treatment approaches, namely cell-based and nanoparticle-based therapies, to overcome the limitations of conventional therapeutics. This paper examines the different facets of each approach, discusses their principles, strengths, and weaknesses, analyses the main targeted pathways and their contradictions, provides insights on current trends as well as highlights any unique mechanisms taken in recent years to combat the progression of atherosclerosis.
Collapse
Affiliation(s)
- Wen Xi Goh
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Yih Yih Kok
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Chiew Yen Wong
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Atherosclerosis severity in patients with familial hypercholesterolemia: The role of T and B lymphocytes. ATHEROSCLEROSIS PLUS 2022; 48:27-36. [PMID: 36644561 PMCID: PMC9833267 DOI: 10.1016/j.athplu.2022.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023]
Abstract
Background and aims Familial hypercholesterolemia (FH) is characterized by lifelong exposure to high LDL-c concentrations and premature atherosclerotic cardiovascular disease; nevertheless, disease severity can be heterogeneous.We aimed at evaluating if the immune-inflammatory system could modulate atherosclerosis burden in FH. Methods From a cohort of subjects with confirmed FH (Dutch Lipid Clinic Network and genotype), 92 patients receiving high-intensity lipid-lowering therapy (statin ± ezetimibe) were included. The extension and severity of coronary atherosclerosis was assessed by standardized reporting systems (CAD-RADS) for coronary computed tomography angiography (CCTA) and coronary artery calcium (CAC) scores. Lipids, apolipoproteins, anti-oxLDL and anti-apolipoprotein B-D peptide (anti-ApoB-D) autoantibodies (IgM and IgG), lymphocytes subtypes, platelet, monocyte and endothelial microparticles (MP), IgM levels (circulating or produced by B1 cells) and cytokines in the supernatant of cultured cells were determined. Multiple linear regression models evaluated associations of these biomarkers with CAC and CAD-RADS scores. Results In univariate analysis CAC correlated with age, systolic blood pressure, TCD4+ cells, and titers of IgM anti-ApoB-D. In multiple linear regression [ANOVA F = 2.976; p = 0.024; R2 = 0.082), CD4+T lymphocytes (B = 35.289; beta = 0.277; p = 0.010; 95%CI for B 8.727 to 61.851), was independently associated with CAC. CAD-RADS correlated with age, systolic blood pressure, titers of IgM anti-ApoB-D, and endothelial MP in univariate analysis. In multiple linear regression, [ANOVA F = 2.790; p = 0.032; R2 = 0.119), only age (B = 0.027; beta = 0.234; p = 0.049; 95% CI for B 0.000 to 0.053) was independent predictor. Conclusions In subjects with FH, under high-intensity lipid-lowering therapy, age and CD4+T cells were associated to atherosclerosis burden.
Collapse
|
4
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
5
|
Understanding Accelerated Atherosclerosis in Systemic Lupus Erythematosus: Toward Better Treatment and Prevention. Inflammation 2021; 44:1663-1682. [PMID: 33821395 DOI: 10.1007/s10753-021-01455-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) carries a significant risk of cardiovascular disease (CVD). The prevalence of premature CVD is especially noteworthy because it occurs in premenopausal women with SLE who would otherwise have very low rates of CVD. While traditional risk factors likely play a role in development of CVD in the setting of SLE, they do not fully explain the excess risk. The pathogenesis of CVD in SLE is not fully understood, but the inflammatory nature of SLE is believed to be a key factor in accelerating atherosclerosis. Systemic inflammation may lead to an abnormal lipid profile with elevated triglycerides, total cholesterol, and low-density lipoprotein cholesterol and dysfunctional high-density lipoprotein cholesterol. Additionally, the inflammatory milieu of SLE plasma promotes endothelial dysfunction and vascular injury, early steps in the progression of atherosclerotic CVD. Despite the overall headway that has been achieved in treating lupus, innovative therapeutics specifically targeting the progression of atherosclerosis within the lupus population are currently lacking. However, there have been advancements in the development of promising modalities for diagnosis of subclinical atherosclerosis and detection of high CVD risk patients. Due to the significant impact of CVD on morbidity and mortality, research addressing prevention and treatment of CVD in SLE needs to be prioritized. This review explores the intricate interplay of SLE-specific properties that contribute to atherosclerosis and CVD within this population, as well as screening methods and possible therapies.
Collapse
|
6
|
Role of B lymphocytes in the infarcted mass in patients with acute myocardial infarction. Biosci Rep 2021; 41:227675. [PMID: 33495783 PMCID: PMC7859321 DOI: 10.1042/bsr20203413] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/05/2021] [Accepted: 01/25/2021] [Indexed: 11/17/2022] Open
Abstract
Despite early reperfusion, patients with ST segment elevation myocardial infarction (STEMI) may present large myocardial necrosis and significant impairment of ventricular function. The present study aimed to evaluate the role of subtypes of B lymphocytes and related cytokines in the infarcted mass and left ventricular ejection fraction obtained by cardiac magnetic resonance imaging performed after 30 days of STEMI. This prospective study included 120 subjects with STEMI submitted to pharmacoinvasive strategy. Blood samples were collected in subjects in the first (D1) and 30th (D30) days post STEMI. The amount of CD11b+ B1 lymphocytes (cells/ml) at D1 were related to the infarcted mass (rho = 0.43; P=0.033), measured by cardiac MRI at D30. These B1 cells were associated with CD4+ T lymphocytes at D1 and D30, while B2 classic lymphocytes at day 30 were related to left ventricular ejection fraction (LVEF). Higher titers of circulating IL-4 and IL-10 were observed at D30 versus D1 (P=0.013 and P<0.001, respectively). Titers of IL-6 at D1 were associated with infarcted mass (rho = 0.41, P<0.001) and inversely related to LVEF (rho = −0.38, P<0.001). After multiple linear regression analysis, high-sensitivity troponin T and IL-6 collected at day 1 were independent predictors of infarcted mass and, at day 30, only HDL-C. Regarding LVEF, high-sensitivity troponin T and high-sensitivity C-reactive protein were independent predictors at day 1, and B2 classic lymphocytes, at day 30. In subjects with STEMI, despite early reperfusion, the amount of infarcted mass and ventricular performance were related to inflammatory responses triggered by circulating B lymphocytes.
Collapse
|
7
|
Zhang S, Zhang S, Lin Z, Zhang X, Dou X, Zhou X, Wang X, Wang Z, Zhang Q. Deep sequencing reveals the skewed B-cell receptor repertoire in plaques and the association between pathogens and atherosclerosis. Cell Immunol 2020; 360:104256. [PMID: 33360167 DOI: 10.1016/j.cellimm.2020.104256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/11/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
The immune/inflammatory responses regulated by B cells are the critical determinants of atherosclerosis. B-cell receptor (BCR) plays pivotal roles in regulating B cell function. However, the composition and molecular characteristics of the BCR repertoire in atherosclerotic patients have not been fully elucidated. Herein we analyzed BCR repertoire in circulation and plaques of atherosclerotic patients by sequencing the BCR heavy chain complement determining region 3 (BCRH CDR3). Our data showed that in plaques, BCR repertoire was dramatically skewed and their combinations and diversity were significantly decreased, while the frequency of public and dominant B-cell clones was markedly increased. Additionally, BCRH CDR3 in plaques had higher positive selection pressure than that in the peripheral blood of normal subjects and atherosclerotic patients. Moreover, the BCRH CDR3 of some B cell clones specifically expanded in plaques were similar to that of antibodies which recognized certain pathogens including Influenza A virus, implying the possibility of the association between pathogens and atherosclerosis. The present study contributed to understand the roles of B cells in atherosclerosis. The design of specific antibodies based on the B cell clones specifically expanded in plaques might yield useful tools to reveal the pathogenesis of atherosclerosis, assess or alleviate the progression of atherosclerosis.
Collapse
Affiliation(s)
- Shucui Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shigang Zhang
- Neurosurgical Department, Liaocheng People's Hospital, Liaocheng, China
| | - Zongwei Lin
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | | | - Xinyao Dou
- Shandong Experimental High School, Jinan, China
| | - Xiaoming Zhou
- Division of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaowei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhe Wang
- Division of Endocrinology and Metabolism, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Qunye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
8
|
Santiago-Raber ML, Montecucco F, Vuilleumier N, Miteva K, Baptista D, Carbone F, Pagano S, Roth A, Burger F, Mach F, Brandt KJ. Atherosclerotic plaque vulnerability is increased in mouse model of lupus. Sci Rep 2020; 10:18324. [PMID: 33110193 PMCID: PMC7591560 DOI: 10.1038/s41598-020-74579-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 10/05/2020] [Indexed: 01/11/2023] Open
Abstract
Anti-apolipoprotein A-1 (anti-apoA-1 IgG) and anti-double stranded DNA (anti-dsDNA IgG) autoantibodies have been described as mediators of atherogenesis in mice and humans. In the present study, we aim to investigate the association between atherosclerotic parameters, autoantibodies and plaque vulnerability in the context of systemic lupus erythematosus (SLE). We therefore bred a lupus prone-mouse model (Nba2.Yaa mice) with Apoe-/- mice resulting in Apoe-/-Nba2.Yaa mice spontaneously producing anti-apoA-1 IgG antibodies. Although Apoe-/-Nba2.Yaa and Apoe-/- mice subject to a high cholesterol diet displayed similar atherosclerosis lesions size in aortic roots and abdominal aorta, the levels of macrophage and neutrophil infiltration, collagen, MMP-8 and MMP-9 and pro-MMP-9 expression in Apoe-/-Nba2.Yaa mice indicated features of atherosclerotic plaque vulnerability. Even though Apoe-/-Nba2.Yaa mice and Apoe-/- mice had similar lipid levels, Apoe-/-Nba2.Yaa mice showed higher anti-apoA-1 and anti-dsDNA IgG levels. Apoe-/-Nba2.Yaa mice displayed a reduction of the size of the kidney, splenomegaly and lymph nodes (LN) hypertrophy. In addition, anti-apoA-1 and anti-dsDNA IgG increased also in relation with mRNA levels of GATA3, IL-4, Bcl-6 and CD20 in the spleen and aortic arch of Apoe-/-Nba2.Yaa mice. Our data show that although atherosclerosis-lupus-prone Apoe-/-Nba2.Yaa mice did not exhibit exacerbated atherosclerotic lesion size, they did show features of atherosclerotic plaque destabilization in correlation with the increase of pro-atherogenic autoantibodies.
Collapse
Affiliation(s)
- Marie-Laure Santiago-Raber
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- AMAL Therapeutics, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Nicolas Vuilleumier
- Department of Genetic Medicine, Laboratory and Pathology, Geneva University Hospitals, Geneva, Switzerland
- Division of Laboratory Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Federico Carbone
- Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Sabrina Pagano
- Department of Genetic Medicine, Laboratory and Pathology, Geneva University Hospitals, Geneva, Switzerland
- Division of Laboratory Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Karim J Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland.
| |
Collapse
|
9
|
Chen L, Ishigami T, Doi H, Arakawa K, Tamura K. Gut microbiota and atherosclerosis: role of B cell for atherosclerosis focusing on the gut-immune-B2 cell axis. J Mol Med (Berl) 2020; 98:1235-1244. [PMID: 32737524 PMCID: PMC7447622 DOI: 10.1007/s00109-020-01936-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/13/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the leading cause of cardiovascular mortality and morbidity worldwide and is described as a complex disease involving several different cell types and their molecular products. Recent studies have revealed that atherosclerosis arises from a systemic inflammatory process, including the accumulation and activities of various immune cells. However, the immune system is a complicated network made up of many cell types, hundreds of bioactive cytokines, and millions of different antigens, making it challenging to readily define the associated mechanism of atherosclerosis. Nevertheless, we previously reported a potential persistent inflammatory process underlying atherosclerosis development, centered on a pathological humoral immune response between commensal microbes and activated subpopulations of substantial B cells in the vicinity of the arterial adventitia. Accumulating evidence has indicated the importance of gut microbiota in atherosclerosis development. Commensal microbiota are considered important regulators of immunity and metabolism and also to be possible antigenic sources for atherosclerosis development. However, the interplay between gut microbiota and metabolism with regard to the modulation of atherosclerosis-associated immune responses remains poorly understood. Here, we review the mechanisms by which the gut microbiota may influence atherogenesis, with particular focus on humoral immunity and B cells, especially the gut-immune-B2 cell axis. Under high-fat and high-calorie conditions, signals driven by the intestinal microbiota via the TLR signaling pathway cause B2 cells in the spleen to become functionally active and activated B2 cells then modify responses such as antibody production (generation of active antibodies IgG and IgG3), thereby contributing to the development of atherosclerosis. On the other hand, intestinal microbiota also resulted in recruitment and ectopic activation of B2 cells via the TLR signaling pathway in perivascular adipose tissue (PVAT), and, subsequently, an increase in circulating IgG and IgG3 led to the enhanced disease development. This is a potential link between microbiota alterations and B cells in the context of atherosclerosis. ![]()
Collapse
Affiliation(s)
- Lin Chen
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.,Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Long Mian Avenue 109 Jiangning, Nanjing, Jiangsu, China
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.
| | - Hiroshi Doi
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Kentaro Arakawa
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
10
|
He D, Mao A, Zheng CB, Kan H, Zhang K, Zhang Z, Feng L, Ma X. Aortic heterogeneity across segments and under high fat/salt/glucose conditions at the single-cell level. Natl Sci Rev 2020; 7:881-896. [PMID: 34692110 PMCID: PMC8289085 DOI: 10.1093/nsr/nwaa038] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 12/24/2022] Open
Abstract
The aorta, with ascending, arch, thoracic and abdominal segments, responds to the heartbeat, senses metabolites and distributes blood to all parts of the body. However, the heterogeneity across aortic segments and how metabolic pathologies change it are not known. Here, a total of 216 612 individual cells from the ascending aorta, aortic arch, and thoracic and abdominal segments of mouse aortas under normal conditions or with high blood glucose levels, high dietary salt, or high fat intake were profiled using single-cell RNA sequencing. We generated a compendium of 10 distinct cell types, mainly endothelial (EC), smooth muscle (SMC), stromal and immune cells. The distributions of the different cells and their intercommunication were influenced by the hemodynamic microenvironment across anatomical segments, and the spatial heterogeneity of ECs and SMCs may contribute to differential vascular dilation and constriction that were measured by wire myography. Importantly, the composition of aortic cells, their gene expression profiles and their regulatory intercellular networks broadly changed in response to high fat/salt/glucose conditions. Notably, the abdominal aorta showed the most dramatic changes in cellular composition, particularly involving ECs, fibroblasts and myeloid cells with cardiovascular risk factor-related regulons and gene expression networks. Our study elucidates the nature and range of aortic cell diversity, with implications for the treatment of metabolic pathologies.
Collapse
Affiliation(s)
- Dongxu He
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Hao Kan
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhiming Zhang
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
11
|
Hosseini H, Yi L, Kanellakis P, Cao A, Tay C, Peter K, Bobik A, Toh BH, Kyaw T. Anti-TIM-1 Monoclonal Antibody (RMT1-10) Attenuates Atherosclerosis By Expanding IgM-producing B1a Cells. J Am Heart Assoc 2018; 7:JAHA.117.008447. [PMID: 29936416 PMCID: PMC6064881 DOI: 10.1161/jaha.117.008447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Peritoneal B1a cells attenuate atherosclerosis by secreting natural polyclonal immunoglobulin M (IgM). Regulatory B cells expressing T‐cell immunoglobulin mucin domain‐1 (TIM‐1) expanded through TIM‐1 ligation by anti‐TIM‐1 monoclonal antibody (RMT1‐10) induces immune tolerance. Methods and Results We examined the capacity of RMT1‐10 to expand peritoneal B1a cells to prevent atherosclerosis development and retard progression of established atherosclerosis. RMT1‐10 treatment selectively doubled peritoneal B1a cells, tripled TIM‐1+ B1a cells and increased TIM‐1+IgM+interleukin (IL)‐10+ by 3‐fold and TIM‐1+IgM+IL‐10− B1a cells by 2.5‐fold. Similar expansion of B1a B cells was observed in spleens. These effects reduced atherosclerotic lesion size, increased plasma IgM and lesion IgM deposits, and decreased oxidatively modified low‐density lipoproteins in lesions. Lesion CD4+ and CD8+ T cells, macrophages and monocyte chemoattractant protein‐1, vascular cell adhesion molecule‐1, expression of proinflammatory cytokines monocyte chemoattractant protein‐1, vascular cell adhesion molecule‐1, IL1β, apoptotic cell numbers and necrotic cores were also reduced. RMT1‐10 treatment failed to expand peritoneal B1a cells and reduce atherosclerosis after splenectomy that reduces B1a cells, indicating that these effects are B1a cell‐dependent. Apolipoprotein E‐KO mice fed a high‐fat diet for 6 weeks before treatment with RMT1‐10 also increased TIM‐1+IgM+IL‐10+ and TIM‐1+IgM+IL‐10− B1a cells and IgM levels and attenuated progression of established atherosclerosis. Conclusions RMT1‐10 treatment attenuates atherosclerosis development and progression by selectively expanding IgM producing atheroprotective B1a cells. Antibody‐based in vivo expansion of B1a cells could be an attractive approach for treating atherosclerosis.
Collapse
Affiliation(s)
- Hamid Hosseini
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | - Li Yi
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | | | - Anh Cao
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Christopher Tay
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | | | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Immunology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | - Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Australia .,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
12
|
Theodorou E, Nezos A, Antypa E, Ioakeimidis D, Koutsilieris M, Tektonidou M, Moutsopoulos HM, Mavragani CP. B-cell activating factor and related genetic variants in lupus related atherosclerosis. J Autoimmun 2018; 92:87-92. [PMID: 29859654 DOI: 10.1016/j.jaut.2018.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease with an increased atherosclerotic risk compared to healthy population, partially explained by traditional cardiovascular (CV) risk factors. Recent data suggest B-cell activating factor (BAFF) as an important contributor in the pathogenesis of both SLE and atherosclerosis. The aim of the current study is to explore whether serum BAFF levels along with variants of the BAFF gene increase lupus related atherosclerotic risk. PATIENTS-METHODS 250 SLE patients underwent assessment of plaque formation and/or intimal media thickness (IMT) measurements in carotid and femoral arteries by ultrasound. Disease related features and CV traditional risk factors were also assessed. Serum BAFF levels were determined by commercially available ELISA and five single nucleotide polymorphisms (SNPs) of the BAFF gene (rs1224141, rs12583006, rs9514828, rs1041569 and the rs9514827) were evaluated by PCR-based assays in all patients and 200 healthy controls (HC) of similar age and sex distribution. SLE patients were further divided in high and low BAFF groups on the basis of the upper quartile level of the distribution (1358 pg/ml). Genotype and haplotype frequencies in SLE patients and HC were determined by SNPStats and SHEsis software. RESULTS High-BAFF SLE group displayed increased rates of both plaque formation and arterial wall thickening (defined as IMT>0.90 mm) compared to patients with low BAFF levels (58.1% vs 43.6%, p:0.048 and 38.6% vs 23.2%, p-value: 0.024, respectively). The association remained significant after disease related features were taken into account (ORs [95%CI]: 2.2 [1.0-5.1] and 2.5 [1.1-5.5] for plaque formation and arterial wall thickening, respectively). Moreover, the presence of the AA genotype of the rs12583006 BAFF gene variant increased susceptibility for both lupus and lupus related plaque formation (ORs [95%CI]: 2.8 [1.1-7.1], and 4.4 [1.3-15.4] in the codominant model, respectively). Finally, the haplotype TTTAT was found to be protective for plaque formation among SLE patients (OR 0.3 [0.1-0.9]. No associations between BAFF gene variants with arterial wall thickening were detected. CONCLUSIONS High BAFF serum levels in the upper 4th quartile as well as BAFF genetic variants seem to increase susceptibility for both lupus and lupus related subclinical atherosclerosis implying B-cell hyperactivity as a potential contributor in the pronounced lupus related atherosclerotic risk.
Collapse
Affiliation(s)
- Evangelos Theodorou
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Rheumatology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Adrianos Nezos
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Antypa
- Department of Radiology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Dimitrios Ioakeimidis
- Department of Rheumatology, General Hospital of Athens "G.Gennimatas", Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Tektonidou
- Rheumatology Unit, First Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Haralampos M Moutsopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio P Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
13
|
Winkels H, Ehinger E, Vassallo M, Buscher K, Dinh HQ, Kobiyama K, Hamers AAJ, Cochain C, Vafadarnejad E, Saliba AE, Zernecke A, Pramod AB, Ghosh AK, Anto Michel N, Hoppe N, Hilgendorf I, Zirlik A, Hedrick CC, Ley K, Wolf D. Atlas of the Immune Cell Repertoire in Mouse Atherosclerosis Defined by Single-Cell RNA-Sequencing and Mass Cytometry. Circ Res 2018; 122:1675-1688. [PMID: 29545366 DOI: 10.1161/circresaha.117.312513] [Citation(s) in RCA: 401] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/04/2018] [Accepted: 03/14/2018] [Indexed: 12/24/2022]
Abstract
RATIONALE Atherosclerosis is a chronic inflammatory disease that is driven by the interplay of pro- and anti-inflammatory leukocytes in the aorta. Yet, the phenotypic and transcriptional diversity of aortic leukocytes is poorly understood. OBJECTIVE We characterized leukocytes from healthy and atherosclerotic mouse aortas in-depth by single-cell RNA-sequencing and mass cytometry (cytometry by time of flight) to define an atlas of the immune cell landscape in atherosclerosis. METHODS AND RESULTS Using single-cell RNA-sequencing of aortic leukocytes from chow diet- and Western diet-fed Apoe-/- and Ldlr-/- mice, we detected 11 principal leukocyte clusters with distinct phenotypic and spatial characteristics while the cellular repertoire in healthy aortas was less diverse. Gene set enrichment analysis on the single-cell level established that multiple pathways, such as for lipid metabolism, proliferation, and cytokine secretion, were confined to particular leukocyte clusters. Leukocyte populations were differentially regulated in atherosclerotic Apoe-/- and Ldlr-/- mice. We confirmed the phenotypic diversity of these clusters with a novel mass cytometry 35-marker panel with metal-labeled antibodies and conventional flow cytometry. Cell populations retrieved by these protein-based approaches were highly correlated to transcriptionally defined clusters. In an integrated screening strategy of single-cell RNA-sequencing, mass cytometry, and fluorescence-activated cell sorting, we detected 3 principal B-cell subsets with alterations in surface markers, functional pathways, and in vitro cytokine secretion. Leukocyte cluster gene signatures revealed leukocyte frequencies in 126 human plaques by a genetic deconvolution strategy. This approach revealed that human carotid plaques and microdissected mouse plaques were mostly populated by macrophages, T-cells, and monocytes. In addition, the frequency of genetically defined leukocyte populations in carotid plaques predicted cardiovascular events in patients. CONCLUSIONS The definition of leukocyte diversity by high-dimensional analyses enables a fine-grained analysis of aortic leukocyte subsets, reveals new immunologic mechanisms and cell-type-specific pathways, and establishes a functional relevance for lesional leukocytes in human atherosclerosis.
Collapse
Affiliation(s)
- Holger Winkels
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Erik Ehinger
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Melanie Vassallo
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Konrad Buscher
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Huy Q Dinh
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Kouji Kobiyama
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Anouk A J Hamers
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.)
| | - Ehsan Vafadarnejad
- Helmholtz Institute for RNA-based Infection Research, Würzburg, Germany (E.V., A.-E.S.)
| | | | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.)
| | - Akula Bala Pramod
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Amlan K Ghosh
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Nathaly Anto Michel
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Natalie Hoppe
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Andreas Zirlik
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.).,Department of Bioengineering, University of California, San Diego (K.L.)
| | - Dennis Wolf
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.) .,From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.).,Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| |
Collapse
|
14
|
Teixeira V, Tam LS. Novel Insights in Systemic Lupus Erythematosus and Atherosclerosis. Front Med (Lausanne) 2018; 4:262. [PMID: 29435447 PMCID: PMC5796914 DOI: 10.3389/fmed.2017.00262] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/28/2017] [Indexed: 01/22/2023] Open
Abstract
Introduction The systemic inflammatory nature of systemic lupus erythematosus (SLE) is well patent not only in the diverse clinical manifestations of the disease but also in the increased risk of premature atherosclerosis and cardiovascular events (CVE), making SLE one of the most complex diseases to study and manage in clinical practice. Aim To travel from old aspects to modern insights on the physiopathology, new molecular biomarkers, imaging methods of atherosclerosis assessment, and the potential treatments of atherosclerosis in SLE. Methods We conducted a literature search using PubMed database and performed a critical review. Conclusion/discussion Several developments have taken place in the understanding of the relationship between SLE and premature atherosclerosis. Nevertheless, cardiovascular diseases are still the major cause of reduced life expectancy in SLE and the main cause of death. The lack of standardization methods for the imaging assessment of atherosclerosis in SLE and the multifactorial nature of the disease are well patriated in the difficulty of achieving consistent and reproducible results among studies that focus in cardiovascular risk assessment and prediction. A raising number of molecular biomarkers of atherosclerosis have been proposed, but the combination of several biomarkers and risk factors may better estimate cardiovascular disease risk. Moreover, the development of effective therapies to prevent progression of atherosclerosis and CVE shall address systemic inflammation.
Collapse
Affiliation(s)
- Vítor Teixeira
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon, Portugal
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
15
|
Abstract
Atherosclerosis is initiated by cholesterol entry into arteries that triggers chronic immune-inflammatory lesions in the vessels. Early lesions are clinically insignificant but advanced complex lesions and vulnerable rupture prone lesions impact on quality of life and can be life threatening. Rupture of vulnerable atherosclerotic lesions initiates thrombotic occlusion of vital arteries precipitating heart attacks and strokes that remain major killers globally despite therapeutic use of statins to lower blood cholesterol levels. Conventional B2 cells are proatherogenic whereas peritoneal Bla cells are atheroprotective. Depletion of B2 cells by administration of mAb to CD20 or to BAFF receptor or in BAFF receptor-deficient mice ameliorates atherosclerosis. B2 cells may promote atherosclerosis by production of IgG, secretion of proinflammatory cytokine TNFα and activation of CD4 T cells. Together these B2 cell mechanisms contribute to generation of rupture-prone vulnerable atherosclerotic plaques characterised by large necrotic cores. In contrast, peritoneal Bla cells protect against atherosclerosis by secretion of natural IgM that scavenges apoptotic cells and oxidised LDL and reduces necrotic cores in atherosclerotic lesions. These atheroprotective effects can be further increased by stimulating Bla cells by administration of apoptotic cells, liposomes of phosphatidylserine abundant on surfaces of apoptotic cell, by mAb to TIM1, a phosphatidylserine receptor expressed by B1a cells and by TLR4-MyD88 activation. Experimental studies of atherosclerosis in mouse models indicate that reductions in atherogenic B2 cells and/or activation of atheroprotective B1a cells protects against atherosclerosis development, findings which have potential for clinical translation to reduce risks of deaths from heart attacks and strokes.
Collapse
Affiliation(s)
- Tin Kyaw
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Peter Tipping
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Alex Bobik
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,c Department of Immunology , Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University , Victoria , Australia
| | - Ban-Hock Toh
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| |
Collapse
|
16
|
Abstract
Angiotensin II (AngII) promotes hypertension, atherogenesis, vascular aneurysm and impairs post-ischemic cardiac remodeling through concerted roles on vascular cells, monocytes and T lymphocytes. However, the role of AngII in B lymphocyte responses is largely unexplored. Here, we show that chronic B cell depletion (Baffr deficiency) significantly reduces atherosclerosis in Apoe−/− mice infused with AngII. While adoptive transfer of B cells in Apoe−/−/Baffr−/− mice reversed atheroprotection in the absence of AngII, infusion of AngII in B cell replenished Apoe−/−/Baffr−/− mice unexpectedly prevented the progression of atherosclerosis. Atheroprotection observed in these mice was associated with a significant increase in regulatory CD1dhiCD5+ B cells, which produced high levels of interleukin (IL)-10 (B10 cells). Replenishment of Apoe−/−/Baffr−/− mice with Il10−/− B cells reversed AngII-induced B cell-dependent atheroprotection, thus highlighting a protective role of IL-10+ regulatory B cells in this setting. Transfer of AngII type 1A receptor deficient (Agtr1a−/−) B cells into Apoe−/−/Baffr−/− mice substantially reduced the production of IL-10 by B cells and prevented the AngII-dependent atheroprotective B cell phenotype. Consistent with the in vivo data, AngII synergized with BAFF to induce IL-10 production by B cells in vitro via AngII type 1A receptor. Our data demonstrate a previously unknown synergy between AngII and BAFF in inducing IL-10 production by B cells, resulting in atheroprotection.
Collapse
|
17
|
Ponnuswamy P, Joffre J, Herbin O, Esposito B, Laurans L, Binder CJ, Tedder TF, Zeboudj L, Loyer X, Giraud A, Zhang Y, Tedgui A, Mallat Z, Ait-Oufella H. Angiotensin II synergizes with BAFF to promote atheroprotective regulatory B cells. Sci Rep 2017. [PMID: 28646220 DOI: 10.1038/s41598‐017‐04438‐6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Angiotensin II (AngII) promotes hypertension, atherogenesis, vascular aneurysm and impairs post-ischemic cardiac remodeling through concerted roles on vascular cells, monocytes and T lymphocytes. However, the role of AngII in B lymphocyte responses is largely unexplored. Here, we show that chronic B cell depletion (Baffr deficiency) significantly reduces atherosclerosis in Apoe -/- mice infused with AngII. While adoptive transfer of B cells in Apoe -/- /Baffr -/- mice reversed atheroprotection in the absence of AngII, infusion of AngII in B cell replenished Apoe -/- /Baffr -/- mice unexpectedly prevented the progression of atherosclerosis. Atheroprotection observed in these mice was associated with a significant increase in regulatory CD1dhiCD5+ B cells, which produced high levels of interleukin (IL)-10 (B10 cells). Replenishment of Apoe -/- /Baffr -/- mice with Il10 -/- B cells reversed AngII-induced B cell-dependent atheroprotection, thus highlighting a protective role of IL-10+ regulatory B cells in this setting. Transfer of AngII type 1A receptor deficient (Agtr1a -/-) B cells into Apoe -/- /Baffr -/- mice substantially reduced the production of IL-10 by B cells and prevented the AngII-dependent atheroprotective B cell phenotype. Consistent with the in vivo data, AngII synergized with BAFF to induce IL-10 production by B cells in vitro via AngII type 1A receptor. Our data demonstrate a previously unknown synergy between AngII and BAFF in inducing IL-10 production by B cells, resulting in atheroprotection.
Collapse
Affiliation(s)
- Padmapriya Ponnuswamy
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Jeremie Joffre
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Olivier Herbin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Bruno Esposito
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Ludivine Laurans
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Christoph J Binder
- Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences and Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas F Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Lynda Zeboudj
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Xavier Loyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Andreas Giraud
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Yujiao Zhang
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Alain Tedgui
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Ziad Mallat
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, England, UK
| | - Hafid Ait-Oufella
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France. .,Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Université Pierre-et-Marie-Curie, Paris, France.
| |
Collapse
|
18
|
Rademakers T, van der Vorst EPC, Daissormont ITMN, Otten JJT, Theodorou K, Theelen TL, Gijbels M, Anisimov A, Nurmi H, Lindeman JHN, Schober A, Heeneman S, Alitalo K, Biessen EAL. Adventitial lymphatic capillary expansion impacts on plaque T cell accumulation in atherosclerosis. Sci Rep 2017; 7:45263. [PMID: 28349940 PMCID: PMC5368662 DOI: 10.1038/srep45263] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/07/2017] [Indexed: 02/07/2023] Open
Abstract
During plaque progression, inflammatory cells progressively accumulate in the adventitia, paralleled by an increased presence of leaky vasa vasorum. We here show that next to vasa vasorum, also the adventitial lymphatic capillary bed is expanding during plaque development in humans and mouse models of atherosclerosis. Furthermore, we investigated the role of lymphatics in atherosclerosis progression. Dissection of plaque draining lymph node and lymphatic vessel in atherosclerotic ApoE-/- mice aggravated plaque formation, which was accompanied by increased intimal and adventitial CD3+ T cell numbers. Likewise, inhibition of VEGF-C/D dependent lymphangiogenesis by AAV aided gene transfer of hVEGFR3-Ig fusion protein resulted in CD3+ T cell enrichment in plaque intima and adventitia. hVEGFR3-Ig gene transfer did not compromise adventitial lymphatic density, pointing to VEGF-C/D independent lymphangiogenesis. We were able to identify the CXCL12/CXCR4 axis, which has previously been shown to indirectly activate VEGFR3, as a likely pathway, in that its focal silencing attenuated lymphangiogenesis and augmented T cell presence. Taken together, our study not only shows profound, partly CXCL12/CXCR4 mediated, expansion of lymph capillaries in the adventitia of atherosclerotic plaque in humans and mice, but also is the first to attribute an important role of lymphatics in plaque T cell accumulation and development.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Emiel P C van der Vorst
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Isabelle T M N Daissormont
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Jeroen J T Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Kosta Theodorou
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Thomas L Theelen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Marion Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Andrey Anisimov
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Harri Nurmi
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Jan H N Lindeman
- Departments of Vascular Surgery and Transplantation Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Schober
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sylvia Heeneman
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Kari Alitalo
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen, Germany
| |
Collapse
|
19
|
Hosseini H, Li Y, Kanellakis P, Tay C, Cao A, Liu E, Peter K, Tipping P, Toh BH, Bobik A, Kyaw T. Toll-Like Receptor (TLR)4 and MyD88 are Essential for Atheroprotection by Peritoneal B1a B Cells. J Am Heart Assoc 2016; 5:e002947. [PMID: 27930350 PMCID: PMC5210362 DOI: 10.1161/jaha.115.002947] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 10/05/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND We previously identified peritoneal B1a cells that secrete natural IgM as a key atheroprotective B cell subset. However, the molecules that activate atheroprotective B1a cells are unknown. Here, we investigated whether Toll-like receptors (TLRs) TLR2, TLR4, and TLR9 expressed by B1a cells are required for IgM-mediated atheroprotection. METHODS AND RESULTS We adoptively transferred B1a cells from wild-type mice or from mice deficient in TLR2, TLR4, TLR9, or myeloid differentiation primary response 88 (MyD88) into ApoE-/- mice depleted of peritoneal B1a cells by splenectomy and fed a high-fat diet for 8 weeks. Elevations in plasma total, anti-oxLDL (oxidized low-density lipoprotein), anti-leukocyte, anti-CD3, anti-CD8, and anti-CD4 IgMs in atherosclerotic mice required B1a cells expressing TLR4 and MyD88, indicating a critical role for TLR4-MyD88 signaling for IgM secretion. Suppression of atherosclerosis was also critically dependent on B1a cells expressing TLR4-MyD88. Atherosclerosis suppression was associated not only with reductions in lesion apoptotic cells, necrotic cores, and oxLDL, but also with reduced lesion CD4+ and CD8+ T cells. Transforming growth factor beta 1 (TGF-β1) expression, including macrophages expressing TGF-β1, was increased, consistent with increased IgM-mediated phagocytosis of apoptotic cells by macrophages. Reductions in lesion inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin (IL) 1β, and IL-18 were consistent with augmented TGF-β1 expression. CONCLUSIONS TLR4-MyD88 expression on B1a cells is critical for their IgM-dependent atheroprotection that not only reduced lesion apoptotic cells and necrotic cores, but also decreased CD4 and CD8 T-cell infiltrates and augmented TGF-β1 expression accompanied by reduced lesion inflammatory cytokines TNF-α, IL-1β, and IL-18.
Collapse
Affiliation(s)
- Hamid Hosseini
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Yi Li
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | | | - Christopher Tay
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Anh Cao
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Edgar Liu
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Karlheinz Peter
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Peter Tipping
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Ban-Hock Toh
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Alex Bobik
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Tin Kyaw
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| |
Collapse
|
20
|
Absence of Activation-induced Cytidine Deaminase, a Regulator of Class Switch Recombination and Hypermutation in B Cells, Suppresses Aorta Allograft Vasculopathy in Mice. Transplantation 2015; 99:1598-605. [PMID: 25769064 DOI: 10.1097/tp.0000000000000688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antibody-mediated rejection is caused in part by increasing circulation/production of donor-specific antibody (DSA). Activation-induced cytidine deaminase (AID) is a key regulator of class switch recombination and somatic hypermutation of immunoglobulin in B cells, yet its role in antibody-mediated transplant rejection remains unclear. We show here that AID deficiency in mice enables suppression of allograft vasculopathy (AV) after aorta transplantation, a DSA-mediated process. METHODS Splenocytes from C57BL/6 J (B6) AID(−/−) mice were used for determining in vitro proliferation responses, alloreactivity, cell surface marker expression, and antibody production. BALB/c mouse aortas were transplanted into B6 AID(−/−) mice with or without FK506 treatment. Blood and aorta grafts were harvested on day 30 after transplantation and were subjected to DSA, histological, and immunohistological analyses. RESULTS The AID(−/−) splenocytes were comparable to wild type splenocytes in proliferation responses, alloreactivity, and expression of cell surface markers in vitro. However, they completely failed to produce immunoglobulin G, although they were not impaired in immunoglobulin M production relative to controls. Furthermore, BALB/c aorta grafts from B6 AID(−/−) recipient mice on day 30 after transplantation showed reduced signs of AV compared to the grafts from B6 wild type recipient mice which had severe vascular intimal hyperplasia, interstitial fibrosis, and inflammation. Treatment with FK506 produced a synergistic effect in the grafts from AID(−/−) recipients with further reduction of intimal hyperplasia and fibrosis scores. CONCLUSIONS The AID deficiency inhibits DSA-mediated AV after aorta transplantation in mice. We propose that AID could be a novel molecular target for controlling antibody-mediated rejection in organ transplantation.
Collapse
|
21
|
Hovland A, Jonasson L, Garred P, Yndestad A, Aukrust P, Lappegård KT, Espevik T, Mollnes TE. The complement system and toll-like receptors as integrated players in the pathophysiology of atherosclerosis. Atherosclerosis 2015; 241:480-94. [PMID: 26086357 DOI: 10.1016/j.atherosclerosis.2015.05.038] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/08/2015] [Accepted: 05/29/2015] [Indexed: 02/08/2023]
Abstract
Despite recent medical advances, atherosclerosis is a global burden accounting for numerous deaths and hospital admissions. Immune-mediated inflammation is a major component of the atherosclerotic process, but earlier research focus on adaptive immunity has gradually switched towards the role of innate immunity. The complement system and toll-like receptors (TLRs), and the crosstalk between them, may be of particular interest both with respect to pathogenesis and as therapeutic targets in atherosclerosis. Animal studies indicate that inhibition of C3a and C5a reduces atherosclerosis. In humans modified LDL-cholesterol activate complement and TLRs leading to downstream inflammation, and histopathological studies indicate that the innate immune system is present in atherosclerotic lesions. Moreover, clinical studies have demonstrated that both complement and TLRs are upregulated in atherosclerotic diseases, although interventional trials have this far been disappointing. However, based on recent research showing an intimate interplay between complement and TLRs we propose a model in which combined inhibition of both complement and TLRs may represent a potent anti-inflammatory therapeutic approach to reduce atherosclerosis.
Collapse
Affiliation(s)
- Anders Hovland
- Coronary Care Unit, Division of Internal Medicine, Nordland Hospital, 8092 Bodø, Norway; Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway.
| | - Lena Jonasson
- Department of Medical and Health Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631 Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Arne Yndestad
- Research Institute of Internal Medicine and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway
| | - Knut T Lappegård
- Coronary Care Unit, Division of Internal Medicine, Nordland Hospital, 8092 Bodø, Norway; Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway
| | - Terje Espevik
- Norwegian University of Science and Technology, Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, 7491 Trondheim, Norway
| | - Tom E Mollnes
- Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway; Norwegian University of Science and Technology, Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, 7491 Trondheim, Norway; Research Laboratory, Nordland Hospital, 8092 Bodø, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0372 Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9019 Tromsø, Norway
| |
Collapse
|
22
|
Hosseini H, Li Y, Kanellakis P, Tay C, Cao A, Tipping P, Bobik A, Toh BH, Kyaw T. Phosphatidylserine liposomes mimic apoptotic cells to attenuate atherosclerosis by expanding polyreactive IgM producing B1a lymphocytes. Cardiovasc Res 2015; 106:443-452. [DOI: 10.1093/cvr/cvv037] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
23
|
Ketelhuth DFJ, Hansson GK. Modulation of autoimmunity and atherosclerosis - common targets and promising translational approaches against disease. Circ J 2015; 79:924-33. [PMID: 25766275 DOI: 10.1253/circj.cj-15-0167] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall that is influenced by several risk factors, including hyperlipidemia and hypertension. Autoimmune diseases substantially increase the risk for cardiovascular disease (CVD). Although atherosclerotic CVD, such as myocardial and stroke, is much more prevalent than classical autoimmune conditions such as rheumatoid arthritis, psoriasis, and systemic lupus erythematosus, these types of pathology have many similarities, raising the possibility that therapies against autoimmune disease can have beneficial effects on CVD. Substantial clinical and experimental data support the potential for immunomodulatory approaches to combating both autoimmune and cardiovascular diseases, including classical immunosuppressants, anticytokine therapy, the targeting of T and B cells and their responses, and vaccination. In this review, we discuss experimental and clinical studies that have used immunomodulatory approaches to mitigate autoimmune reactions and examine their potential to prevent and treat atherosclerotic CVD.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital
| | | |
Collapse
|
24
|
Role of vascular inflammation in coronary artery disease: potential of anti-inflammatory drugs in the prevention of atherothrombosis. Inflammation and anti-inflammatory drugs in coronary artery disease. Am J Cardiovasc Drugs 2015; 15:1-11. [PMID: 25369900 DOI: 10.1007/s40256-014-0094-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronary artery disease (CAD) and acute myocardial infarction (AMI) are inflammatory pathologies, involving interleukins (ILs), such as IL-1β, IL-6 and tumor necrosis factor (TNF)-α, and acute phase proteins production, such as for C reactive protein (CRP). The process begins with retention of low-density lipoprotein (LDL) and its oxidation inside the intima, with the formation of the "foam cells." Toll-like receptors and inflamassomes participate in atherosclerosis formation, as well as in the activation of the complement system. In addition to innate immunity, adaptive immunity is also associated with atherosclerosis through antigen-presenting cells, T and B lymphocytes. AMI also increases the expression of some ILs and promotes macrophage and lymphocyte accumulation. Reperfusion increases the expression of anti-inflammatory ILs (such as IL-10) and generates oxygen free radicals. Although CAD and AMI are inflammatory disorders, the only drugs with anti-inflammatory effect so far widely used in ischemic heart disease are aspirin and statins. Some immunomodulatory or immunosuppressive promising therapies, such as cyclosporine and colchicine, may have benefits in CAD. Methotrexate also has potential cardioprotective anti-inflammatory effects, through increased adenosine levels. The TETHYS trial (The Effects of mETHotrexate Therapy on ST Segment Elevation MYocardial InfarctionS trial) will evaluate low-dose methotrexate in ST elevation AMI. The CIRT (Cardiovascular Inflammation Reduction Trial), in turn, will evaluate low-dose methotrexate in patients with a high prevalence of subclinical vascular inflammation. The CANTOS (The Canakinumab Antiinflammatory Thrombosis Outcomes Study) will evaluate canakinumab in patients with CAD and persistently elevated CRP. The blockage of other potential targets, such as the IL-6 receptor, CC2 chemokine receptor and CD20, could bring benefits in CAD.
Collapse
|
25
|
Tsiantoulas D, Sage AP, Mallat Z, Binder CJ. Targeting B cells in atherosclerosis: closing the gap from bench to bedside. Arterioscler Thromb Vasc Biol 2014; 35:296-302. [PMID: 25359862 DOI: 10.1161/atvbaha.114.303569] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Atherosclerotic plaque formation is strongly influenced by different arms of the immune system, including B lymphocytes. B cells are divided into 2 main families: the B1 and the B2 cells. B1 cells are atheroprotective mainly via the production of natural IgM antibodies that bind oxidized low-density lipoprotein and apoptotic cells. B2 cells, which include follicular and marginal zone B cells, are suggested to be proatherogenic. Antibody-mediated depletion of B cells has become a valuable treatment option for certain autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis that are also characterized by the development of premature atherosclerosis. Thus, B cells represent a novel interesting target for therapeutic modulation of the atherosclerotic disease process. Here, we discuss the effect of different of B-cell subsets in experimental atherosclerosis, their mechanism of action as well as potential ways to exploit these findings for the treatment of human disease.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Andrew P Sage
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Ziad Mallat
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Christoph J Binder
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.).
| |
Collapse
|
26
|
|
27
|
Gjurich BN, Taghavie-Moghadam PL, Ley K, Galkina EV. L-selectin deficiency decreases aortic B1a and Breg subsets and promotes atherosclerosis. Thromb Haemost 2014; 112:803-11. [PMID: 24989887 DOI: 10.1160/th13-10-0865] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
There is a significant recruitment of leucocytes into aortas during atherogenesis. L-selectin regulates leucocyte migration into secondary lymphoid and peripheral tissues and was proposed to play a role in leucocyte homing into aortas. Here, we determine the role of L-selectin in atherosclerosis. L-selectin-deficient Apoe-/- (Sell-/-Apoe-/-) mice had a 74% increase in plaque burden compared to Apoe-/- mice fed a chow diet for 50 weeks. Elevated atherosclerosis was accompanied by increased aortic leucocyte content, but a 50% reduction in aortic B cells despite elevated B cell counts in the blood. Follicular B cells represented 65%, whereas B1a and regulatory B cells (Breg) comprised 5% of aortic B cells. B1a and Breg cell subsets were reduced in Sell-/-Apoe-/- aortas with accompanied two-fold decrease in aortic T15 antibody and 1.2-fold decrease of interleukin-10 (IL-10) levels. L-selectin was required for B1 cell homing to the atherosclerotic aorta, as demonstrated by a 1.5-fold decrease in the migration of Sell-/-Apoe-/- vs Apoe-/- cells. Notably, we found a 1.6-fold increase in CD68hi macrophages in Sell-/-Apoe-/- compared to Apoe-/- aortas, despite comparable blood monocyte numbers and L-selectin-dependent aortic homing. L-selectin had no effect on neutrophil migration into aorta, but led to elevated blood neutrophil numbers, suggesting a potential involvement of neutrophils in atherogenesis of Sell-/-Apoe-/- mice. Thus, L-selectin deficiency increases peripheral blood neutrophil and lymphocyte numbers, decreases aortic B1a and Breg populations, T15 antibody and IL-10 levels, and increases aortic macrophage content of Sell-/-Apoe-/- mice. Altogether, these data provide evidence for an overall atheroprotective role of L-selectin.
Collapse
Affiliation(s)
| | | | | | - Elena V Galkina
- Elena V. Galkina, PhD, Associate Professor, Dept. Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA 23507-1696, USA, Tel.: +1 757 446 5019, Fax: +1 757 624 2255, E-mail:
| |
Collapse
|
28
|
Libby P, Tabas I, Fredman G, Fisher EA. Inflammation and its resolution as determinants of acute coronary syndromes. Circ Res 2014; 114:1867-79. [PMID: 24902971 PMCID: PMC4078767 DOI: 10.1161/circresaha.114.302699] [Citation(s) in RCA: 389] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/12/2014] [Indexed: 12/28/2022]
Abstract
Inflammation contributes to many of the characteristics of plaques implicated in the pathogenesis of acute coronary syndromes. Moreover, inflammatory pathways not only regulate the properties of plaques that precipitate acute coronary syndromes but also modulate the clinical consequences of the thrombotic complications of atherosclerosis. This synthesis will provide an update on the fundamental mechanisms of inflammatory responses that govern acute coronary syndromes and also highlight the ongoing balance between proinflammatory mechanisms and endogenous pathways that can promote the resolution of inflammation. An appreciation of the countervailing mechanisms that modulate inflammation in relation to acute coronary syndromes enriches our fundamental understanding of the pathophysiology of this important manifestation of atherosclerosis. In addition, these insights provide glimpses into potential novel therapeutic interventions to forestall this ultimate complication of the disease.
Collapse
Affiliation(s)
- Peter Libby
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.).
| | - Ira Tabas
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| | - Gabrielle Fredman
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| | - Edward A Fisher
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| |
Collapse
|
29
|
Klasen C, Ohl K, Sternkopf M, Shachar I, Schmitz C, Heussen N, Hobeika E, Levit-Zerdoun E, Tenbrock K, Reth M, Bernhagen J, El Bounkari O. MIF promotes B cell chemotaxis through the receptors CXCR4 and CD74 and ZAP-70 signaling. THE JOURNAL OF IMMUNOLOGY 2014; 192:5273-84. [PMID: 24760155 DOI: 10.4049/jimmunol.1302209] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with chemokine-like functions that plays a pivotal role in the pathogenesis of inflammatory diseases by promoting leukocyte recruitment. We showed that MIF promotes the atherogenic recruitment of monocytes and T cells through its receptors CXCR2 and CXCR4. Effects of MIF on B cell recruitment have not been addressed. In this study, we tested the involvement of MIF in B cell chemotaxis and studied the underlying mechanism. We show that MIF promotes primary murine B cell chemotaxis in a dose-dependent manner, comparable to the B cell chemokines CXCL13 and CXCL12. Splenic B cells express CXCR4 and the receptor CD74 but not CXCR2. Inhibition of CXCR4 or CD74 or a genetic deficiency of Cd74 in primary B cells fully abrogated MIF-mediated B cell migration, implying cooperative involvement of both receptors. MIF stimulation of B cells resulted in a rapid increase in intracellular Ca(2+) mobilization and F-actin polymerization. Intriguingly, the tyrosine kinase ZAP-70 was activated upon MIF and CXCL12 treatment in a CXCR4- and CD74-dependent manner. Pharmacological inhibition of ZAP-70 resulted in abrogation of primary B cell migration. Functional involvement of ZAP-70 was confirmed by small interfering RNA-mediated knockdown in Ramos B cell migration. Finally, primary B cells from ZAP-70 gene-deficient mice exhibited ablated transmigration in response to MIF or CXCL12. We conclude that MIF promotes the migration of B cells through a ZAP-70-dependent pathway mediated by cooperative engagement of CXCR4 and CD74. The data also suggest that MIF may contribute to B cell recruitment in vivo (e.g., in B cell-related immune disorders).
Collapse
Affiliation(s)
- Christina Klasen
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | - Kim Ohl
- Department of Pediatrics, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | - Marieke Sternkopf
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | | | - Corinna Schmitz
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | - Nicole Heussen
- Institute of Medical Statistics, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | - Elias Hobeika
- Department of Molecular Immunology, Biology III, University of Freiburg, 79108 Freiburg, Germany; and Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Ella Levit-Zerdoun
- Department of Molecular Immunology, Biology III, University of Freiburg, 79108 Freiburg, Germany; and Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany
| | - Michael Reth
- Department of Molecular Immunology, Biology III, University of Freiburg, 79108 Freiburg, Germany; and Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany;
| | - Omar El Bounkari
- Institute of Biochemistry and Molecular Cell Biology, Rheinisch-Westfälisch Technical University (RWTH) Aachen, D-52074 Aachen, Germany;
| |
Collapse
|
30
|
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
|
31
|
Gaudreault N, Kumar N, Olivas VR, Eberlé D, Stephens K, Raffai RL. Hyperglycemia impairs atherosclerosis regression in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1981-1992. [PMID: 24113453 DOI: 10.1016/j.ajpath.2013.08.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/23/2013] [Accepted: 08/12/2013] [Indexed: 02/01/2023]
Abstract
Diabetic patients are known to be more susceptible to atherosclerosis and its associated cardiovascular complications. However, the effects of hyperglycemia on atherosclerosis regression remain unclear. We hypothesized that hyperglycemia impairs atherosclerosis regression by modulating the biological function of lesional macrophages. HypoE (Apoe(h/h)Mx1-Cre) mice express low levels of apolipoprotein E (apoE) and develop atherosclerosis when fed a high-fat diet. Atherosclerosis regression occurs in these mice upon plasma lipid lowering induced by a change in diet and the restoration of apoE expression. We examined the morphological characteristics of regressed lesions and assessed the biological function of lesional macrophages isolated with laser-capture microdissection in euglycemic and hyperglycemic HypoE mice. Hyperglycemia induced by streptozotocin treatment impaired lesion size reduction (36% versus 14%) and lipid loss (38% versus 26%) after the reversal of hyperlipidemia. However, decreases in lesional macrophage content and remodeling in both groups of mice were similar. Gene expression analysis revealed that hyperglycemia impaired cholesterol transport by modulating ATP-binding cassette A1, ATP-binding cassette G1, scavenger receptor class B family member (CD36), scavenger receptor class B1, and wound healing pathways in lesional macrophages during atherosclerosis regression. Hyperglycemia impairs both reduction in size and loss of lipids from atherosclerotic lesions upon plasma lipid lowering without significantly affecting the remodeling of the vascular wall.
Collapse
Affiliation(s)
- Nathalie Gaudreault
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California
| | - Nikit Kumar
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California
| | - Victor R Olivas
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California
| | - Delphine Eberlé
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California
| | - Kyle Stephens
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California
| | - Robert L Raffai
- Surgical Service, VA Medical Center San Francisco, San Francisco, California; Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California.
| |
Collapse
|
32
|
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: 225] [Impact Index Per Article: 18.8] [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
|
33
|
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]
|
34
|
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
|
35
|
Leonard D, Svenungsson E, Sandling JK, Berggren O, Jönsen A, Bengtsson C, Wang C, Jensen-Urstad K, Granstam SO, Bengtsson AA, Gustafsson JT, Gunnarsson I, Rantapää-Dahlqvist S, Nordmark G, Eloranta ML, Syvänen AC, Rönnblom L. Coronary heart disease in systemic lupus erythematosus is associated with interferon regulatory factor-8 gene variants. ACTA ACUST UNITED AC 2013; 6:255-63. [PMID: 23661672 DOI: 10.1161/circgenetics.113.000044] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Patients with systemic lupus erythematosus have increased morbidity and mortality in coronary heart disease (CHD). We asked whether there was a genetic influence on CHD in systemic lupus erythematosus. METHODS AND RESULTS The association between single-nucleotide polymorphisms (SNPs) and CHD in 2 populations of patients with systemic lupus erythematosus was assessed. Patients were genotyped on a custom 12k Illumina Array. The allele frequencies were compared between patients with (n=66) and without (n=509) CHD. We found 61 SNPs with an association (P<0.01) to CHD, with the strongest association for 3 SNPs located in the interferon regulatory factor-8 (IRF8) gene. Comparison of the allele frequencies of these 61 SNPs in patients with (n=27) and without (n=212) CHD in the second study population revealed that 2 SNPs, rs925994 and rs10514610 in IRF8 (linkage disequilibrium, r²=0.84), were associated with CHD in both study populations. Meta-analysis of the SNP rs925994 gave an odds ratio of 3.6 (2.1-6.3), P value 1.9×10⁻⁶. The identified IRF8 allele remained as a risk factor for CHD after adjustment for traditional CHD risk factors. The IRF8 risk allele was associated with the presence of carotid plaques (P<0.001) and increased intima-media thickness (P=0.01). By electrophoretic mobility shift assays, we show weaker binding of protein to the risk allele of the highly linked SNP rs11117415, and by flow cytometry, a reduced frequency of circulating B cells was detected in patients with the IRF8 risk allele. CONCLUSIONS There is a considerable genetic component for CHD in systemic lupus erythematosus, with IRF8 as a strong susceptibility locus.
Collapse
Affiliation(s)
- Dag Leonard
- Section of Rheumatology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Carbone F, Nencioni A, Mach F, Vuilleumier N, Montecucco F. Evidence on the pathogenic role of auto-antibodies in acute cardiovascular diseases. Thromb Haemost 2013; 109:854-868. [PMID: 23446994 DOI: 10.1160/th12-10-0768] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/29/2013] [Indexed: 02/06/2023]
Abstract
Atherothrombosis is the major determinant of acute ischaemic cardiovascular events, such as myocardial infarction and stroke. Inflammatory processes have been linked to all phases of atherogenesis In particular, the identification of autoimmunity mediators in the complex microenvironment of chronic inflammation has become the focus of attention in both early and advanced atherogenic processes. Auto-antibodies against self-molecules or new epitopes generated by oxidative processes infiltrate atherosclerotic plaques and were shown to modulate the activity of immune cells by binding various types of receptors. However, despite mounting evidence for a pathophysiological role of autoantibodies in atherothrombosis, the clinical relevance for circulating autoantibodies in cardiovascular outcomes is still debated. This review aims at illustrating the mechanisms by which different types of autoantibodies might either promote or repress atherothrombosis and to discuss the clinical studies assessing the role of auto-antibodies as prognostic biomarkers of plaque vulnerability.
Collapse
Affiliation(s)
- F Carbone
- Cardiology Division, Department of Medicine, Geneva University Hospital, Foundation for Medical Researches, 64 Avenue Roseraie, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
37
|
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: 71] [Impact Index Per Article: 5.9] [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
|
38
|
Shao W, Jin H, Huang J, Qiu B, Xia R, Deng Z, Cai J, Chen Y. AFM investigation on Ox-LDL-induced changes in cell spreading and cell-surface adhesion property of endothelial cells. SCANNING 2013; 35:119-126. [PMID: 22833475 DOI: 10.1002/sca.21040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 06/16/2012] [Indexed: 06/01/2023]
Abstract
The integrity and adhesion properties of endothelium play vital roles during atherosclerosis. It is well known that oxidized low-density lipoprotein (Ox-LDL) influences many physiological activities or mechanical properties of endothelial cells. However, the effects of Ox-LDL on the integrity and nonspecific adhesion properties of endothelial cells are still unclear. In this study, using the topographical imaging and force measurement functions of atomic force microscopy (AFM), we found that Ox-LDL can transiently weaken the integrity of endothelium by impairing cell spreading of endothelial cells and decrease the attachment of irrelevant blood cells to endothelium by impairing the nonspecific adhesion property of endothelial cells. The AFM-based data provide important information for understanding the effects of Ox-LDL on endothelial cells or during atherogenesis.
Collapse
Affiliation(s)
- Wenxiang Shao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science 2013; 339:161-6. [PMID: 23307733 DOI: 10.1126/science.1230719] [Citation(s) in RCA: 833] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases claim more lives worldwide than any other. Etiologically, the dominant trajectory involves atherosclerosis, a chronic inflammatory process of lipid-rich lesion growth in the vascular wall that can cause life-threatening myocardial infarction (MI). Those who survive MI can develop congestive heart failure, a chronic condition of inadequate pump activity that is frequently fatal. Leukocytes (white blood cells) are important participants at the various stages of cardiovascular disease progression and complication. This Review will discuss leukocyte function in atherosclerosis, MI, and heart failure.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA.
| | | |
Collapse
|
40
|
Ishizaka N. IgG4-related disease underlying the pathogenesis of coronary artery disease. Clin Chim Acta 2013; 415:220-5. [DOI: 10.1016/j.cca.2012.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 10/30/2012] [Accepted: 11/02/2012] [Indexed: 01/05/2023]
|
41
|
Regulation of atherogenesis by chemokines and chemokine receptors. Arch Immunol Ther Exp (Warsz) 2012; 61:1-14. [PMID: 23224338 DOI: 10.1007/s00005-012-0202-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 11/18/2012] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is a chronic inflammatory and metabolic disorder affecting large- and medium-sized arteries, and the leading cause of mortality worldwide. The pathogenesis of atherosclerosis involves accumulation of lipids and leukocytes in the intima of blood vessel walls creating plaque. How leukocytes accumulate in plaque remains poorly understood; however, chemokines acting at specific G protein-coupled receptors appear to be important. Studies using knockout mice suggest that chemokine receptor signaling may either promote or inhibit atherogenesis, depending on the receptor. These proof of concept studies have spurred efforts to develop drugs targeting the chemokine system in atherosclerosis, and several have shown beneficial effects in animal models. This study will review key discoveries in basic and translational research in this area.
Collapse
|
42
|
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]
|
43
|
Role of Peroxisome Proliferator-Activated Receptor-γ in Vascular Inflammation. Int J Vasc Med 2012; 2012:508416. [PMID: 22888436 PMCID: PMC3409528 DOI: 10.1155/2012/508416] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 06/08/2012] [Indexed: 12/13/2022] Open
Abstract
Vascular inflammation plays a crucial role in atherosclerosis, and its regulation is important to prevent cerebrovascular and coronary artery disease. The inflammatory process in atherogenesis involves a variety of immune cells including monocytes/macrophages, lymphocytes, dendritic cells, and neutrophils, which all express peroxisome proliferator-activated receptor-γ (PPAR-γ). PPAR-γ is a nuclear receptor and transcription factor in the steroid superfamily and is known to be a key regulator of adipocyte differentiation. Increasing evidence from mainly experimental studies has demonstrated that PPAR-γ activation by endogenous and synthetic ligands is involved in lipid metabolism and anti-inflammatory activity. In addition, recent clinical studies have shown a beneficial effect of thiazolidinediones, synthetic PPAR-γ ligands, on cardiovascular disease beyond glycemic control. These results suggest that PPAR-γ activation is an important regulator in vascular inflammation and is expected to be a therapeutic target in the treatment of atherosclerotic complications. This paper reviews the recent findings of PPAR-γ involvement in vascular inflammation and the therapeutic potential of regulating the immune system in atherosclerosis.
Collapse
|
44
|
Sage AP, Tsiantoulas D, Baker L, Harrison J, Masters L, Murphy D, Loinard C, Binder CJ, Mallat Z. BAFF Receptor Deficiency Reduces the Development of Atherosclerosis in Mice—Brief Report. Arterioscler Thromb Vasc Biol 2012; 32:1573-6. [DOI: 10.1161/atvbaha.111.244731] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
The goal of this study was to assess the role of B-cell activating factor (BAFF) receptor in B-cell regulation of atherosclerosis.
Methods and Results—
Male LDL receptor-deficient mice (
Ldlr
−/−
) were lethally irradiated and reconstituted with either wild type or BAFF receptor (BAFF-R)–deficient bone marrow. After 4 weeks of recovery, mice were put on a high-fat diet for 6 or 8 weeks. BAFF-R deficiency in bone marrow cells led to a marked reduction of conventional mature B2 cells but did not affect the B1a cell subtype. This was associated with a significant reduction of dendritic cell activation and T-cell proliferation along with a reduction of IgG antibodies against malondialdehyde-modified low-density lipoprotein. In contrast, serum IgM type antibodies were preserved. Interestingly, BAFF-R deficiency was associated with a significant reduction in atherosclerotic lesion development and reduced numbers of plaque T cells. Selective BAFF-R deficiency on B cells led to a similar reduction in lesion size and T-cell infiltration but in contrast did not affect dendritic cell activation.
Conclusion—
BAFF-R deficiency in mice selectively alters mature B2 cell-dependent cellular and humoral immune responses and limits the development of atherosclerosis.
Collapse
Affiliation(s)
- Andrew P. Sage
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Lauren Baker
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - James Harrison
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Leanne Masters
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Deirdre Murphy
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Celine Loinard
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Christoph J. Binder
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine (A.P.S., L.B., J.H., L.M., D.M., C.L., Z.M.), University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK; Center for Molecular Medicine (D.T., C.J.B.) of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine (D.T., C.J.B.), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Campbell KA, Lipinski MJ, Doran AC, Skaflen MD, Fuster V, McNamara CA. Lymphocytes and the adventitial immune response in atherosclerosis. Circ Res 2012; 110:889-900. [PMID: 22427326 DOI: 10.1161/circresaha.111.263186] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although much of the research on atherosclerosis has focused on the intimal accumulation of lipids and inflammatory cells, there is an increasing amount of interest in the role of the adventitia in coordinating the immune response in atherosclerosis. In this review of the contributions of the adventitia and adventitial lymphocytes to the development of atherosclerosis, we discuss recent research on the formation and structural nature of adventitial immune aggregates, potential mechanisms of crosstalk between the intima, media, and adventitia, specific contributions of B lymphocytes and T lymphocytes, and the role of the vasa vasorum and surrounding perivascular adipose tissue. Furthermore, we highlight techniques for the imaging of lymphocytes in the vasculature.
Collapse
Affiliation(s)
- Kirsti A Campbell
- Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | | | | | | | | | | |
Collapse
|
46
|
Skaggs BJ, Hahn BH, McMahon M. Accelerated atherosclerosis in patients with SLE--mechanisms and management. Nat Rev Rheumatol 2012; 8:214-23. [PMID: 22331061 PMCID: PMC3765069 DOI: 10.1038/nrrheum.2012.14] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rapid-onset cardiovascular disease (CVD) is a major concern for many patients with systemic lupus erythematosus (SLE). Cardiovascular events occur more frequently and with earlier onset in patients with SLE compared with healthy individuals. Traditional risk factors, such as altered lipid levels, aging and smoking, do not fully explain this increased risk of CVD, strongly suggesting that autoimmunity contributes to accelerated atherosclerosis. Altered immune system function is recognized as the primary contributor to both the initiation and progression of atherosclerosis. Multiple manifestations of autoimmunity, including changes in cytokine levels and innate immune responses, autoantibodies, adipokines, dysfunctional lipids, and oxidative stress, could heighten atherosclerotic risk. In addition, multiple SLE therapeutics seem to affect the development and progression of atherosclerosis both positively and negatively. SLE-specific cardiovascular risk factors are beginning to be discovered by several groups, and development of a comprehensive, clinically feasible biomarker panel could be invaluable for identification and treatment of patients at risk of developing accelerated atherosclerosis. Here, we discuss the epidemiology of CVD in SLE and the implications of immune system dysfunction on the development and progression, monitoring and treatment of atherosclerosis in individuals with this disease.
Collapse
Affiliation(s)
- Brian J Skaggs
- Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, 32-59 Rehab Center, 1000 Veteran Avenue, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
47
|
Kyaw T, Tay C, Hosseini H, Kanellakis P, Gadowski T, MacKay F, Tipping P, Bobik A, Toh BH. Depletion of B2 but not B1a B cells in BAFF receptor-deficient ApoE mice attenuates atherosclerosis by potently ameliorating arterial inflammation. PLoS One 2012; 7:e29371. [PMID: 22238605 PMCID: PMC3251583 DOI: 10.1371/journal.pone.0029371] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/27/2011] [Indexed: 11/18/2022] Open
Abstract
We have recently identified conventional B2 cells as atherogenic and B1a cells as atheroprotective in hypercholesterolemic ApoE−/− mice. Here, we examined the development of atherosclerosis in BAFF-R deficient ApoE−/− mice because B2 cells but not B1a cells are selectively depleted in BAFF-R deficient mice. We fed BAFF-R−/− ApoE−/− (BaffR.ApoE DKO) and BAFF-R+/+ApoE−/− (ApoE KO) mice a high fat diet (HFD) for 8-weeks. B2 cells were significantly reduced by 82%, 81%, 94%, 72% in blood, peritoneal fluid, spleen and peripheral lymph nodes respectively; while B1a cells and non-B lymphocytes were unaffected. Aortic atherosclerotic lesions assessed by oil red-O stained-lipid accumulation and CD68+ macrophage accumulation were decreased by 44% and 50% respectively. B cells were absent in atherosclerotic lesions of BaffR.ApoE DKO mice as were IgG1 and IgG2a immunoglobulins produced by B2 cells, despite low but measurable numbers of B2 cells and IgG1 and IgG2a immunoglobulin concentrations in plasma. Plasma IgM and IgM deposits in atherosclerotic lesions were also reduced. BAFF-R deficiency in ApoE−/− mice was also associated with a reduced expression of VCAM-1 and fewer macrophages, dendritic cells, CD4+ and CD8+ T cell infiltrates and PCNA+ cells in lesions. The expression of proinflammatory cytokines, TNF-α, IL1-β and proinflammatory chemokine MCP-1 was also reduced. Body weight and plasma cholesterols were unaffected in BaffR.ApoE DKO mice. Our data indicate that B2 cells are important contributors to the development of atherosclerosis and that targeting the BAFF-R to specifically reduce atherogenic B2 cell numbers while preserving atheroprotective B1a cell numbers may be a potential therapeutic strategy to reduce atherosclerosis by potently reducing arterial inflammation.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, Victoria, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Atherosclerosis is a complex vascular pathology characterized in part by accumulation of innate and adaptive inflammatory cells in arterial plaque. Molecular mediators responsible for inflammatory cell accumulation in plaque include specific members of the chemokine family of leukocyte chemoattractants and their G protein-coupled receptors. Studies using the ApoE knockout mouse model have recently implicated chemokine receptor Ccr6 and its ligand Ccl20 as a nonredundant ligand-receptor pair in atherosclerosis, potentially operating at several stages of cell recruitment and on several leukocyte subtypes.
Collapse
Affiliation(s)
- Wuzhou Wan
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|