1
|
Zhang L, Li P, Li Y, Qu W, Shi Y, Zhang T, Chen Y. The role of immunoglobins in atherosclerosis development; friends or foe? Mol Cell Biochem 2025; 480:2737-2747. [PMID: 39592554 DOI: 10.1007/s11010-024-05158-y] [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: 06/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Coronary artery disease, atherosclerosis, and its life-threatening sequels impose the hugest burden on the healthcare systems throughout the world. The intricate process of atherosclerosis is considered as an inflammatory-based disorder, and therefore, the components of the immune system are involved in different stages from formation of coronary plaques to its development. One of the major effectors in this way are the antibody producing entities, the B cells. These cells, which play a significant and unique role in responding to different stress, injuries, and infections, contribute differently to the development of atherosclerosis, either inhibitory or promoting, depending on the type of subsets. B cells implicate in both systemic and local immune responses of an atherosclerotic artery by cell-cell contact, cytokine production, and antigen presentation. In particular, natural antibodies bind to oxidized lipoproteins and cellular debris, which are abundant during plaque growth. Logically, any defects in B cells and consequent impairment in antibody production may greatly affect the shaping of the plaque and its clinical outcome. In this comprehensive review, we scrutinize the role of B cells and different classes of antibodies in atherosclerosis progression besides current novel B-cell-based therapeutic approaches that aim to resolve this affliction of mankind.
Collapse
Affiliation(s)
- Linlin Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Peize Li
- Department of Orthopedics, Changchun Chinese Medicine Hospital, Changchun, 130022, China
| | - Yuhui Li
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Wantong Qu
- Department of Cardiology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yanyu Shi
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Tianyang Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ying Chen
- Department of Cardiology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
2
|
Dedemadi AG, Gkolfinopoulou C, Nikoleri D, Nikoloudaki M, Ruhanen H, Holopainen M, Käkelä R, Christopoulou G, Bournazos S, Constantoulakis P, Sidiropoulos P, Bertsias G, Chroni A. Improvement of high-density lipoprotein atheroprotective properties in patients with systemic lupus erythematosus after belimumab treatment. Rheumatology (Oxford) 2025; 64:648-657. [PMID: 38514392 PMCID: PMC11781589 DOI: 10.1093/rheumatology/keae192] [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: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
OBJECTIVE Chronic inflammatory diseases, like Systemic Lupus Erythematosus (SLE), carry an increased risk for atherosclerosis and cardiovascular events, accompanied by impairment of atheroprotective properties of high-density lipoprotein (HDL). In SLE, serum B cell-activating factor (BAFF), a cytokine implicated in disease progression, has been correlated with subclinical atherosclerosis. We investigated the impact of treatment with belimumab -an anti-BAFF monoclonal antibody- on HDL atheroprotective properties and composition in SLE patients. METHODS Serum samples were collected from 35 SLE patients with active disease despite conventional therapy, before and after 6-month add-on treatment with belimumab, and 26 matched healthy individuals. We measured cholesterol efflux and antioxidant capacities, paraoxonase-1 (PON1) activity, serum amyloid A1 (SAA1), myeloperoxidase (MPO) and lipid peroxidation product levels of HDL. LC-MS/MS was performed to analyse the HDL lipidome. RESULTS Following treatment with belimumab, cholesterol efflux and antioxidant capacities of HDL were significantly increased in SLE patients and restored to levels of control subjects. HDL-associated PON1 activity was also increased, whereas lipid peroxidation products were decreased following treatment. HDL cholesterol efflux and antioxidant capacities correlated negatively with the disease activity. Changes were noted in the HDL lipidome of SLE patients following belimumab treatment, as well as between SLE patients and healthy individuals, and specific changes in lipid species correlated with functional parameters of HDL. CONCLUSIONS HDL of SLE patients with active disease displays impaired atheroprotective properties accompanied by distinct lipidomic signatures compared with controls. Belimumab treatment may improve the HDL atheroprotective properties and modify the HDL lipidomic signature in SLE patients, thus potentially mitigating atherosclerosis development.
Collapse
Affiliation(s)
- Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Athens, Greece
| | - Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
| | - Dimitra Nikoleri
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Myrto Nikoloudaki
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Minna Holopainen
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | | | | | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - George Bertsias
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
| |
Collapse
|
3
|
Giakomidi D, Ishola A, Nus M. Targeting gut microbiota to regulate the adaptive immune response in atherosclerosis. Front Cardiovasc Med 2025; 12:1502124. [PMID: 39957996 PMCID: PMC11825770 DOI: 10.3389/fcvm.2025.1502124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the leading cause of death worldwide, is a chronic inflammatory disease leading to the accumulation of lipid-rich plaques in the intima of large and medium-sized arteries. Accumulating evidence indicates the important regulatory role of the adaptive immune system in atherosclerosis during all stages of the disease. The gut microbiome has also become a key regulator of atherosclerosis and immunomodulation. Whilst existing research extensively explores the impact of the microbiome on the innate immune system, only a handful of studies have explored the regulatory capacity of the microbiome on the adaptive immune system to modulate atherogenesis. Building on these concepts and the pitfalls on the gut microbiota and adaptive immune response interaction, this review explores potential strategies to therapeutically target the microbiome, including the use of prebiotics and vaccinations, which could influence the adaptive immune response and consequently plaque composition and development.
Collapse
Affiliation(s)
- Despina Giakomidi
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Ayoola Ishola
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Wang W, Xia J, Shen Y, Qiao C, Liu M, Cheng X, Mu S, Yan W, Lu W, Gao S, Zhou K. Potential diagnostic biomarkers in heart failure: Suppressed immune-associated genes identified by bioinformatic analysis and machine learning. Eur J Pharmacol 2025; 986:177153. [PMID: 39586393 DOI: 10.1016/j.ejphar.2024.177153] [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: 07/26/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Heart failure (HF) threatens tens of millions of people's health worldwide, which is the terminal stage in the development of majority cardiovascular diseases. Recently, an increasing number of studies have demonstrated that bioinformatics and machine learning (ML) algorithms can offer new insights into the diagnosing and treating HF. To further discover HF diagnostic genes, we utilized least absolute shrinkage and selection operator (LASSO) and Support Vector Machine (SVM) to identify novel immune-related genes. The HF dataset was obtained from the gene expression omnibus (GEO) database and three candidate genes (LCN6, MUC4, and TNFRSF13C) were finally screened. In addition, the myocardial infarction (MI) modeling experiments on mice were performed to validate the expression of LCN6, MUC4, and TNFRSF13C on experimental HF mice. Altogether, these three candidate genes are promising targets for the prediction of HF with immunological perspective.
Collapse
Affiliation(s)
- Wanrong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jie Xia
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yu Shen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chuncan Qiao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Mengyan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xin Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Siqi Mu
- First Clinical Medical College, Anhui Medical University, Hefei, 230032, China
| | - Weizhen Yan
- Department of Oncology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Wenjie Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Shan Gao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Kai Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China; The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Juhasz V, Charlier FT, Zhao TX, Tsiantoulas D. Targeting the adaptive immune continuum in atherosclerosis and post-MI injury. Atherosclerosis 2024; 399:118616. [PMID: 39546915 DOI: 10.1016/j.atherosclerosis.2024.118616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024]
Abstract
Atherosclerotic disease is a cholesterol-rich lipoprotein particle-driven disease resulting in the formation of atherosclerotic plaques in large and medium size arteries. Rupture or erosion of atherosclerotic plaques can trigger the formation of a thrombus causing the obstruction of the blood flow in the coronary artery and thereby leading to myocardial infarction (MI). Inflammation is a crucial pillar of the mechanisms leading to atherosclerosis and governing the cardiac repair post-MI. Dissecting the complex and sophisticated networks of the immune responses underlying the formation of atherosclerotic plaques and affecting the healing of the heart after MI will allow the designing of highly precise immunomodulatory therapies for these settings. Notably, MI also accelerates atherosclerosis via modulating the response of the immune system. Therefore, for the identification of effective and safe therapeutic targets, it is critical to consider the inflammatory continuum that interconnects the two pathologies and identify immunomodulatory strategies that confer a protective effect in both settings or at least, affect each pathology independently. Adaptive immunity, which consists of B and T lymphocytes, is a major regulator of atherosclerosis and post-MI cardiac repair. Here, we review and discuss the effect of potential adaptive immunity-targeting therapies, such as cell-depleting therapies, in atherosclerosis and post-MI cardiac injury.
Collapse
Affiliation(s)
- Viktoria Juhasz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fiona T Charlier
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | | |
Collapse
|
6
|
Porsch F, Binder CJ. Autoimmune diseases and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:780-807. [PMID: 38937626 DOI: 10.1038/s41569-024-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Autoimmune diseases are associated with a dramatically increased risk of atherosclerotic cardiovascular disease and its clinical manifestations. The increased risk is consistent with the notion that atherogenesis is modulated by both protective and disease-promoting immune mechanisms. Notably, traditional cardiovascular risk factors such as dyslipidaemia and hypertension alone do not explain the increased risk of cardiovascular disease associated with autoimmune diseases. Several mechanisms have been implicated in mediating the autoimmunity-associated cardiovascular risk, either directly or by modulating the effect of other risk factors in a complex interplay. Aberrant leukocyte function and pro-inflammatory cytokines are central to both disease entities, resulting in vascular dysfunction, impaired resolution of inflammation and promotion of chronic inflammation. Similarly, loss of tolerance to self-antigens and the generation of autoantibodies are key features of autoimmunity but are also implicated in the maladaptive inflammatory response during atherosclerotic cardiovascular disease. Therefore, immunomodulatory therapies are potential efficacious interventions to directly reduce the risk of cardiovascular disease, and biomarkers of autoimmune disease activity could be relevant tools to stratify patients with autoimmunity according to their cardiovascular risk. In this Review, we discuss the pathophysiological aspects of the increased cardiovascular risk associated with autoimmunity and highlight the many open questions that need to be answered to develop novel therapies that specifically address this unmet clinical need.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Cao W, Wang K, Wang J, Chen Y, Gong H, Xiao L, Pan W. Causal relationship between immune cells and risk of myocardial infarction: evidence from a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1416112. [PMID: 39257847 PMCID: PMC11384581 DOI: 10.3389/fcvm.2024.1416112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024] Open
Abstract
Background Atherosclerotic plaque rupture is a major cause of heart attack. Previous studies have shown that immune cells are involved in the development of atherosclerosis, but different immune cells play different roles. The aim of this study was to investigate the causal relationship between immunological traits and myocardial infarction (MI). Methods To assess the causal association of immunological profiles with myocardial infarction based on publicly available genome-wide studies, we used a two-sample mendelian randomization (MR) approach with inverse variance weighted (IVW) as the main analytical method. Sensitivity analyses were used to assess heterogeneity and horizontal pleiotropy. Results A two-sample MR analysis was conducted using IVW as the primary method. At a significance level of 0.001, we identified 47 immunophenotypes that have a significant causal relationship with MI. Seven of these were present in B cells, five in cDC, four in T cells at the maturation stage, six in monocytes, five in myeloid cells, 12 in TBNK cells, and eight in Treg cells. Sensitivity analyses were performed to confirm the robustness of the MR results. Conclusions Our results provide strong evidence that multiple immune cells have a causal effect on the risk of myocardial infarction. This discovery provides a new avenue for the development of therapeutic treatments for myocardial infarction and a new target for drug development.
Collapse
Affiliation(s)
- Wenjing Cao
- Cardiology Department, Geriatrics Department, Foshan Women and Children Hospital, Foshan, Guangdong, China
| | - Kui Wang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiawei Wang
- Department of Critical Care Medicine, Jieyang Third People's Hospital, Jieyang, China
| | - Yuhua Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hanxian Gong
- Cardiology Department, Geriatrics Department, Foshan Women and Children Hospital, Foshan, Guangdong, China
| | - Lei Xiao
- Cardiology Department, Geriatrics Department, Foshan Women and Children Hospital, Foshan, Guangdong, China
| | - Wei Pan
- Cardiology Department, Geriatrics Department, Foshan Women and Children Hospital, Foshan, Guangdong, China
| |
Collapse
|
8
|
O’Brien JW, Case A, Kemper C, Zhao TX, Mallat Z. Therapeutic Avenues to Modulate B-Cell Function in Patients With Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2024; 44:1512-1522. [PMID: 38813699 PMCID: PMC11208059 DOI: 10.1161/atvbaha.124.319844] [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] [Indexed: 05/31/2024]
Abstract
The adaptive immune system plays an important role in the development and progression of atherosclerotic cardiovascular disease. B cells can have both proatherogenic and atheroprotective roles, making treatments aimed at modulating B cells important therapeutic targets. The innate-like B-cell response is generally considered atheroprotective, while the adaptive response is associated with mixed consequences for atherosclerosis. Additionally, interactions of B cells with components of the adaptive and innate immune system, including T cells and complement, also represent key points for therapeutic regulation. In this review, we discuss therapeutic approaches based on B-cell depletion, modulation of B-cell survival, manipulation of both the antibody-dependent and antibody-independent B-cell response, and emerging immunization techniques.
Collapse
Affiliation(s)
- James W. O’Brien
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
| | - Ayden Case
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (C.K.)
| | - Tian X. Zhao
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
- Department of Cardiology, Royal Papworth Hospital, Cambridge, United Kingdom (T.X.Z.)
| | - Ziad Mallat
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
- Unversité de Paris, Inserm U970, Paris Cardiovascular Research Centre, France (Z.M.)
| |
Collapse
|
9
|
Obare LM, Bonami RH, Doran A, Wanjalla CN. B cells and atherosclerosis: A HIV perspective. J Cell Physiol 2024; 239:e31270. [PMID: 38651687 PMCID: PMC11209796 DOI: 10.1002/jcp.31270] [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: 12/04/2023] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Atherosclerosis remains a leading cause of cardiovascular disease (CVD) globally, with the complex interplay of inflammation and lipid metabolism at its core. Recent evidence suggests a role of B cells in the pathogenesis of atherosclerosis; however, this relationship remains poorly understood, particularly in the context of HIV. We review the multifaceted functions of B cells in atherosclerosis, with a specific focus on HIV. Unique to atherosclerosis is the pivotal role of natural antibodies, particularly those targeting oxidized epitopes abundant in modified lipoproteins and cellular debris. B cells can exert control over cellular immune responses within atherosclerotic arteries through antigen presentation, chemokine production, cytokine production, and cell-cell interactions, actively participating in local and systemic immune responses. We explore how HIV, characterized by chronic immune activation and dysregulation, influences B cells in the context of atherosclerosis, potentially exacerbating CVD risk in persons with HIV. By examining the proatherogenic and antiatherogenic properties of B cells, we aim to deepen our understanding of how B cells influence atherosclerotic plaque development, especially within the framework of HIV. This research provides a foundation for novel B cell-targeted interventions, with the potential to mitigate inflammation-driven cardiovascular events, offering new perspectives on CVD risk management in PLWH.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amanda Doran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
10
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
11
|
Sanges S, Tian W, Dubucquoi S, Chang JL, Collet A, Launay D, Nicolls MR. B-cells in pulmonary arterial hypertension: friend, foe or bystander? Eur Respir J 2024; 63:2301949. [PMID: 38485150 PMCID: PMC11043614 DOI: 10.1183/13993003.01949-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/01/2024] [Indexed: 04/22/2024]
Abstract
There is an unmet need for new therapeutic strategies that target alternative pathways to improve the prognosis of patients with pulmonary arterial hypertension (PAH). As immunity has been involved in the development and progression of vascular lesions in PAH, we review the potential contribution of B-cells in its pathogenesis and evaluate the relevance of B-cell-targeted therapies. Circulating B-cell homeostasis is altered in PAH patients, with total B-cell lymphopenia, abnormal subset distribution (expansion of naïve and antibody-secreting cells, reduction of memory B-cells) and chronic activation. B-cells are recruited to the lungs through local chemokine secretion, and activated by several mechanisms: 1) interaction with lung vascular autoantigens through cognate B-cell receptors; 2) costimulatory signals provided by T follicular helper cells (interleukin (IL)-21), type 2 T helper cells and mast cells (IL-4, IL-6 and IL-13); and 3) increased survival signals provided by B-cell activating factor pathways. This activity results in the formation of germinal centres within perivascular tertiary lymphoid organs and in the local production of pathogenic autoantibodies that target the pulmonary vasculature and vascular stabilisation factors (including angiotensin-II/endothelin-1 receptors and bone morphogenetic protein receptors). B-cells also mediate their effects through enhanced production of pro-inflammatory cytokines, reduced anti-inflammatory properties by regulatory B-cells, immunoglobulin (Ig)G-induced complement activation, and IgE-induced mast cell activation. Precision-medicine approaches targeting B-cell immunity are a promising direction for select PAH conditions, as suggested by the efficacy of anti-CD20 therapy in experimental models and a trial of rituximab in systemic sclerosis-associated PAH.
Collapse
Affiliation(s)
- Sébastien Sanges
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Wen Tian
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Sylvain Dubucquoi
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - Jason L Chang
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Aurore Collet
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - David Launay
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| | - Mark R Nicolls
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| |
Collapse
|
12
|
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
13
|
Pattarabanjird T, Srikakulapu P, Ransegnola B, Marshall MA, Ghosheh Y, Gulati R, Durant C, Drago F, Taylor AM, Ley K, McNamara CA. Single-cell profiling of CD11c+ B cells in atherosclerosis. Front Immunol 2024; 14:1296668. [PMID: 38259450 PMCID: PMC10800418 DOI: 10.3389/fimmu.2023.1296668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Circulating CD11c+ B cells, a novel subset of activated B cells, have been linked to autoimmunity and shown to expand with age. Atherosclerosis is an age-associated disease that involves innate and adaptive immune responses to modified self-antigens. Yet, the expression of CD11c on specific B-cell subtypes and its link to atherosclerosis are poorly understood. In this study, we characterized the frequency of CD11c+ B cells in tissues in mice with aging. We observed an age-associated increase in CD11c+ B cells in the spleen and bone marrow of ApoE-/- mice, and this was associated with an increase in aortic plaque. In addition, we also utilized single-cell multi-omics profiling of 60 human subjects undergoing advanced imaging for coronary artery disease (CAD) to subtype CD11c+ B cells and determine their frequency in subjects with high and low severity of CAD. Using unsupervised clustering, we identified four distinct clusters of CD11c+ B cells, which include CD27 and IgD double negative 2 (DN2), age-associated (ABC), CD11c+ unswitched memory (USWM), and activated Naïve (aNav) B cells. We observed an increase in the frequency of both ABC B cells and DN2 B cells in patients with high CAD severity. Pathway analysis further demonstrated augmentation of autophagy, IFNg signaling, and TLR signaling in DN2 cells in high-severity CAD patients. On the other hand, an increase in the negative regulator of BCR signaling through CD72 was found in ABC cells in low-severity CAD patients. Through investigating scRNAseq of atheroma, these DN2 cells were also found to infiltrate human coronary atheroma.
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Brett Ransegnola
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Melissa A. Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Yanal Ghosheh
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Rishab Gulati
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Angela M. Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Klaus Ley
- Immunology Center of Georgia, Augusta University, Augusta, GA, United States
| | - Coleen A. McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
14
|
Zhao L, Ma D, Wang L, Su X, Feng L, Zhu L, Chen Y, Hao Y, Wang X, Feng J. Metabolic changes with the occurrence of atherosclerotic plaques and the effects of statins. Front Immunol 2023; 14:1301051. [PMID: 38143759 PMCID: PMC10739339 DOI: 10.3389/fimmu.2023.1301051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Atherosclerosis is a common cardiovascular disease caused by the abnormal expression of multiple factors and genes influenced by both environmental and genetic factors. The primary manifestation of atherosclerosis is plaque formation, which occurs when inflammatory cells consume excess lipids, affecting their retention and modification within the arterial intima. This triggers endothelial cell (EC) activation, immune cell infiltration, vascular smooth muscle cell (VSMC) proliferation and migration, foam cell formation, lipid streaks, and fibrous plaque development. These processes can lead to vascular wall sclerosis, lumen stenosis, and thrombosis. Immune cells, ECs, and VSMCs in atherosclerotic plaques undergo significant metabolic changes and inflammatory responses. The interaction of cytokines and chemokines secreted by these cells leads to the onset, progression, and regression of atherosclerosis. The regulation of cell- or cytokine-based immune responses is a novel therapeutic approach for atherosclerosis. Statins are currently the primary pharmacological agents utilised for managing unstable plaques owing to their ability to enhance endothelial function, regulate VSMC proliferation and apoptosis by reducing cholesterol levels, and mitigate the expression and activity of inflammatory cytokines. In this review, we provide an overview of the metabolic changes associated with atherosclerosis, describe the effects of inflammatory responses on atherosclerotic plaques, and discuss the mechanisms through which statins contribute to plaque stabilisation. Additionally, we examine the role of statins in combination with other drugs in the management of atherosclerosis.
Collapse
Affiliation(s)
| | - Di Ma
- Bethune First Hospital, Jilin University, Changchun, China
| | - LiJuan Wang
- Bethune First Hospital, Jilin University, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
16
|
Li K, Li K, He Y, Liang S, Shui X, Lei W. Aryl hydrocarbon receptor: A bridge linking immuno-inflammation and metabolism in atherosclerosis. Biochem Pharmacol 2023; 216:115744. [PMID: 37579858 DOI: 10.1016/j.bcp.2023.115744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide, and atherosclerosis is a major contributor to this etiology. The ligand-activated transcription factor, known as the aryl hydrocarbon receptor (AhR), plays an essential role in the interactions between genes and the environment. In a number of human diseases, including atherosclerosis, the AhR signaling pathway has recently been shown to be aberrantly expressed and activated. It's reported that AhR can regulate the immuno-inflammatory response and metabolism pathways in atherosclerosis, potentially serving as a bridge that links these processes. In this review, we highlight the involvement of AhR in atherosclerosis. From the literature, we conclude that AhR is a potential target for controlling atherosclerosis through precise interventions.
Collapse
Affiliation(s)
- Kongwei Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kaiyue Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shan Liang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaorong Shui
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
17
|
Wang Q, Xue Q. Bioinformatics analysis of potential common pathogenic mechanism for carotid atherosclerosis and Parkinson's disease. Front Aging Neurosci 2023; 15:1202952. [PMID: 37649719 PMCID: PMC10464527 DOI: 10.3389/fnagi.2023.1202952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background Cerebrovascular disease (CVD) related to atherosclerosis and Parkinson's disease (PD) are two prevalent neurological disorders. They share common risk factors and frequently occur together. The aim of this study is to investigate the association between atherosclerosis and PD using genetic databases to gain a comprehensive understanding of underlying biological mechanisms. Methods The gene expression profiles of atherosclerosis (GSE28829 and GSE100927) and PD (GSE7621 and GSE49036) were downloaded from the Gene Expression Omnibus (GEO) database. After identifying the common differentially expressed genes (DEGs) for these two disorders, we constructed protein-protein interaction (PPI) networks and functional modules, and further identified hub genes using Least Absolute Shrinkage and Selection Operator (LASSO) regression. The diagnostic effectiveness of these hub genes was evaluated using Receiver Operator Characteristic Curve (ROC) analysis. Furthermore, we used single sample gene set enrichment analysis (ssGSEA) to analyze immune cell infiltration and explored the association of the identified hub genes with infiltrating immune cells through Spearman's rank correlation analysis in R software. Results A total of 50 shared DEGs, with 36 up-regulated and 14 down-regulated genes, were identified through the intersection of DEGs of atherosclerosis and PD. Using LASSO regression, we identified six hub genes, namely C1QB, CD53, LY96, P2RX7, C3, and TNFSF13B, in the lambda.min model, and CD14, C1QB, CD53, P2RX7, C3, and TNFSF13B in the lambda.1se model. ROC analysis confirmed that both models had good diagnostic efficiency for atherosclerosis datasets GSE28829 (lambda.min AUC = 0.99, lambda.1se AUC = 0.986) and GSE100927 (lambda.min AUC = 0.922, lambda.1se AUC = 0.933), as well as for PD datasets GSE7621 (lambda.min AUC = 0.924, lambda.1se AUC = 0.944) and GSE49036 (lambda.min AUC = 0.894, lambda.1se AUC = 0.881). Furthermore, we found that activated B cells, effector memory CD8 + T cells, and macrophages were the shared correlated types of immune cells in both atherosclerosis and PD. Conclusion This study provided new sights into shared molecular mechanisms between these two disorders. These common hub genes and infiltrating immune cells offer promising clues for further experimental studies to explore the common pathogenesis of these disorders.
Collapse
Affiliation(s)
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Ebrahimian T, Dierick F, Ta V, Kotsiopriftis M, O'Connor Miranda J, Mann KK, Orthwein A, Lehoux S. B cell-specific knockout of AID protects against atherosclerosis. Sci Rep 2023; 13:8723. [PMID: 37253865 DOI: 10.1038/s41598-023-35980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/26/2023] [Indexed: 06/01/2023] Open
Abstract
Antigen-naive IgM-producing B cells are atheroprotective, whereas mature B cells producing class-switched antibodies promote atherosclerosis. Activation-induced cytidine deaminase (AID), which mediates class switch recombination (CSR), would thus be expected to foster atherosclerosis. Yet, AID also plays a major role in the establishment of B cell tolerance. We sought to define whether AID affects atherosclerotic plaque formation. We generated Ldlr-/- chimeras transplanted with bone marrow from Aicda-/- or wild-type (WT) mice, fed a HFD for 14 weeks. Decreased B cell maturation in Ldlr-/-Aicda-/- mice was demonstrated by 50% reduction in splenic and aortic BAFFR expression, a key signaling component of B2 cell maturation. This was associated with increased plasma IgM in Ldlr-/-Aicda-/- compared with Ldlr-/-WT animals. Importantly, Ldlr-/-Aicda-/- mice had reduced atherosclerotic lesion area (0.20 ± 0.03mm2) compared with Ldlr-/-WT (0.30 ± 0.04mm2, P < 0.05), although no differences in plaque composition were noted between groups. In addition, immunofluorescence analysis revealed increased splenic B and T cell areas independent of cell number. AID depletion directly inhibits atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Talin Ebrahimian
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada.
| | - France Dierick
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Vincent Ta
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Maria Kotsiopriftis
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | | | - Koren K Mann
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Alexandre Orthwein
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Stephanie Lehoux
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada.
| |
Collapse
|
19
|
Taylor JA, Hutchinson MA, Gearhart PJ, Maul RW. Antibodies in action: the role of humoral immunity in the fight against atherosclerosis. Immun Ageing 2022; 19:59. [PMID: 36461105 PMCID: PMC9717479 DOI: 10.1186/s12979-022-00316-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
The sequestering of oxidation-modified low-density lipoprotein by macrophages results in the accumulation of fatty deposits within the walls of arteries. Necrosis of these cells causes a release of intercellular epitopes and the activation of the adaptive immune system, which we predict leads to robust autoantibody production. T cells produce cytokines that act in the plaque environment and further stimulate B cell antibody production. B cells in atherosclerosis meanwhile have a mixed role based on subclass. The current model is that B-1 cells produce protective IgM antibodies in response to oxidation-specific epitopes that work to control plaque formation, while follicular B-2 cells produce class-switched antibodies (IgG, IgA, and IgE) which exacerbate the disease. Over the course of this review, we discuss further the validation of these protective antibodies while evaluating the current dogma regarding class-switched antibodies in atherosclerosis. There are several contradictory findings regarding the involvement of class-switched antibodies in the disease. We hypothesize that this is due to antigen-specificity, and not simply isotype, being important, and that a closer evaluation of these antibodies' targets should be conducted. We propose that specific antibodies may have therapeutical potential in preventing and controlling plaque development within a clinical setting.
Collapse
Affiliation(s)
- Joshua A. Taylor
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Mark A. Hutchinson
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| | - Patricia J. Gearhart
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| | - Robert W. Maul
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| |
Collapse
|
20
|
Martos-Folgado I, del Monte-Monge A, Lorenzo C, Busse CE, Delgado P, Mur SM, Cobos-Figueroa L, Escolà-Gil JC, Martín-Ventura JL, Wardemann H, Ramiro AR. MDA-LDL vaccination induces athero-protective germinal-center-derived antibody responses. Cell Rep 2022; 41:111468. [DOI: 10.1016/j.celrep.2022.111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/13/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
|
21
|
Li GQ, Liu XM, Liu BL, Zhong Y, Gu QW, Miao JJ, Wang J, Liu S, Mao XM. High triiodothyronine levels induce myocardial hypertrophy via BAFF overexpression. J Cell Mol Med 2022; 26:4453-4462. [PMID: 35808902 PMCID: PMC9357614 DOI: 10.1111/jcmm.17470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/18/2022] [Indexed: 11/27/2022] Open
Abstract
Activated B cells contribute to heart diseases, and inhibition of B‐cell activating factor (BAFF) expression is an effective therapeutic target for heart diseases. Whether activated B cells participate in the development and progression of hyperthyroid heart disease, and what induces B cells activation in hyperthyroidism are unknown. The present study aimed to determine the roles of BAFF overexpression induced by high concentrations of triiodothyronine (T3) in the pathogenesis of hyperthyroid heart disease. Female C57BL/6J mice were subcutaneously injected with T3 for 6 weeks, and BAFF expression was inhibited using shRNA. Protein and mRNA expression of BAFF in mouse heart tissues evaluated via immunohistochemistry, western blotting and polymerase chain reaction (PCR). Proportions of B cells in mouse cardiac tissue lymphocytes were quantified via flow cytometry. Morphology and left ventricle function were assessed using pathological sections and echocardiography, respectively. Here, we demonstrate that compared with the control group, the proportion of myocardial B cells was larger in the T3 group; immunohistochemistry, western blotting and PCR analyses revealed increased protein and mRNA expression levels of TNF‐α and BAFF in heart tissues of the T3 group. Compared with the normal controls group, in the T3 group, the diameter of myocardial cells and some echocardiographic values significantly increased and hypertrophy and structural disorder were noticeable. Our results revealed that elevated levels of circulating T3 can promote the expression of BAFF in myocardial cells and can lead to B‐cell activation, an elevated inflammatory response and ventricular remodelling.
Collapse
Affiliation(s)
- Guo-Qing Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Mei Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bing-Li Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Zhong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qing-Wei Gu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jing-Jing Miao
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shu Liu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Ming Mao
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Mallat Z, Binder CJ. The why and how of adaptive immune responses in ischemic cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:431-444. [PMID: 36382200 PMCID: PMC7613798 DOI: 10.1038/s44161-022-00049-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/10/2022] [Indexed: 02/02/2023]
Abstract
Atherosclerotic cardiovascular disease is a major cause of disability and death worldwide. Most therapeutic approaches target traditional risk factors but ignore the fundamental role of the immune system. This is a huge unmet need. Recent evidence indicates that reducing inflammation may limit cardiovascular events. However, the concomitant increase in the risk of lifethreatening infections is a major drawback. In this context, targeting adaptive immunity could constitute a highly effective and safer approach. In this Review, we address the why and how of the immuno-cardiovascular unit, in health and in atherosclerotic disease. We review and discuss fundamental mechanisms that ensure immune tolerance to cardiovascular tissue, and examine how their disruption promotes disease progression. We identify promising strategies to manipulate the adaptive immune system for patient benefit, including novel biologics and RNA-based vaccination strategies. Finally, we advocate for establishing a molecular classification of atherosclerosis as an important milestone in our quest to radically change the understanding and treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
- Unversité de Paris, and INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022; 44:309-324. [PMID: 35355124 PMCID: PMC9064999 DOI: 10.1007/s00281-022-00922-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022. [PMID: 35355124 DOI: 10.1007/s00281-02200922-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
25
|
Smeets D, Gisterå A, Malin SG, Tsiantoulas D. The Spectrum of B Cell Functions in Atherosclerotic Cardiovascular Disease. Front Cardiovasc Med 2022; 9:864602. [PMID: 35497984 PMCID: PMC9051234 DOI: 10.3389/fcvm.2022.864602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
B cells are a core element of the pathophysiology of atherosclerotic cardiovascular disease (ASCVD). Multiple experimental and epidemiological studies have revealed both protective and deleterious functions of B cells in atherosclerotic plaque formation. The spearhead property of B cells that influences the development of atherosclerosis is their unique ability to produce and secrete high amounts of antigen-specific antibodies that can act at distant sites. Exposure to an atherogenic milieu impacts B cell homeostasis, cell differentiation and antibody production. However, it is not clear whether B cell responses in atherosclerosis are instructed by atherosclerosis-specific antigens (ASA). Dissecting the full spectrum of the B cell properties in atherosclerosis will pave the way for designing innovative therapies against the devastating consequences of ASCVD.
Collapse
Affiliation(s)
- Diede Smeets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Anton Gisterå
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Stephen G. Malin
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
26
|
Pattarabanjird T, Marshall M, Upadhye A, Srikakulapu P, Garmey J, Haider A, Taylor AM, Lutgens E, McNamara CA. B-1b Cells Possess Unique bHLH-Driven P62-Dependent Self-Renewal and Atheroprotection. Circ Res 2022; 130:981-993. [PMID: 35209718 PMCID: PMC9075598 DOI: 10.1161/circresaha.121.320436] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND B1a and B1b lymphocytes produce IgM that inactivates oxidation-specific epitopes (IgMOSE) on LDL (low-density lipoprotein) and protects against atherosclerosis. Loss of ID3 (inhibitor of differentiation 3) in B cells selectively promotes B1b but not B1a cell numbers, leading to higher IgMOSE production and reduction in atherosclerotic plaque formation. Yet, the mechanism underlying this regulation remains unexplored. METHODS Bulk RNA sequencing was utilized to identify differentially expressed genes in B1a and B1b cells from Id3KO and Id3WT mice. CRISPR/Cas9 and lentiviral genome editing coupled with adoptive transfer were used to identify key Id3-dependent signaling pathways regulating B1b cell proliferation and the impact on atherosclerosis. Biospecimens from humans with advanced coronary artery disease imaging were analyzed to translate murine findings to human subjects with coronary artery disease. RESULTS Through RNA sequencing, P62 was found to be enriched in Id3KO B1b cells. Further in vitro characterization reveals a novel role for P62 in mediating BAFF (B-cell activating factor)-induced B1b cell proliferation through interacting with TRAF6 and activating NF-κB (nuclear factor kappa B), leading to subsequent C-MYC upregulation. Promoter-reporter assays reveal that Id3 inhibits the E2A protein from activating the P62 promoter. Mice adoptively transferred with B1 cells overexpressing P62 exhibited an increase in B1b cell number and IgMOSE levels and were protected against atherosclerosis. Consistent with murine mechanistic findings, P62 expression in human B1 cells was significantly higher in subjects harboring a function-impairing SNP (rs11574) in the ID3 gene and directly correlated with plasma IgMOSE levels. CONCLUSIONS This study unveils a novel role for P62 in driving BAFF-induced B1b cell proliferation and IgMOSE production to attenuate diet-induced atherosclerosis. Results identify a direct role for Id3 in antagonizing E2A from activating the p62 promoter. Moreover, analysis of putative human B1 cells also implicates these pathways in coronary artery disease subjects, suggesting P62 as a new immunomodulatory target for treating atherosclerosis.
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Melissa Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
| | - Aditi Upadhye
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
| | - Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
| | - James Garmey
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
| | - Antony Haider
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
| | - Angela M. Taylor
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany; and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Coleen A. McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia, United States
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
- Correspondence: Corresponding Author, , Phone: 434-243-5854, Address: 345 Crispell Dr. Charlottesville, VA 22908
| |
Collapse
|
27
|
Douna H, de Mol J, Amersfoort J, Schaftenaar FH, Kiss MG, Suur BE, Kroner MJ, Binder CJ, Bot I, Van Puijvelde GHM, Kuiper J, Foks AC. IFNγ-Stimulated B Cells Inhibit T Follicular Helper Cells and Protect Against Atherosclerosis. Front Cardiovasc Med 2022; 9:781436. [PMID: 35187121 PMCID: PMC8847680 DOI: 10.3389/fcvm.2022.781436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
Abstract
B and T cells are interconnected in the T follicular helper—germinal center B cell (TFH-GC B cell) axis, which is hyperactive during atherosclerosis development and loss of control along this axis results in exacerbated atherosclerosis. Inhibition of the TFH–GC B cell axis can be achieved by providing negative co-stimulation to TFH cells through the PD-1/PD-L1 pathway. Therefore, we investigated a novel therapeutic strategy using PD-L1-expressing B cells to inhibit atherosclerosis. We found that IFNγ-stimulated B cells significantly enhanced PD-L1 expression and limited TFH cell development. To determine whether IFNγ-B cells can reduce collar-induced atherosclerosis, apoE−/− mice fed a Western-type diet were treated with PBS, B cells or IFNγ-B cells for a total of 5 weeks following collar placement. IFNγ-B cells significantly increased PD-L1hi GC B cells and reduced plasmablasts. Interestingly, IFNγ-B cells–treated mice show increased atheroprotective Tregs and T cell-derived IL-10. In line with these findings, we observed a significant reduction in total lesion volume in carotid arteries of IFNγ-B cells-treated mice compared to PBS-treated mice and a similar trend was observed compared to B cell-treated mice. In conclusion, our data show that IFNγ-stimulated B cells strongly upregulate PD-L1, inhibit TFH cell responses and protect against atherosclerosis.
Collapse
Affiliation(s)
- Hidde Douna
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - J. de Mol
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Jacob Amersfoort
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Frank H. Schaftenaar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Mate G. Kiss
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bianca E. Suur
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Mara J. Kroner
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Gijs H. M. Van Puijvelde
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
- *Correspondence: Amanda C. Foks
| |
Collapse
|
28
|
Spinosa MD, Montgomery WG, Lempicki M, Srikakulapu P, Johnsrude MJ, McNamara CA, Upchurch GR, Ailawadi G, Leitinger N, Meher AK. B Cell-Activating Factor Antagonism Attenuates the Growth of Experimental Abdominal Aortic Aneurysm. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2231-2244. [PMID: 34509440 PMCID: PMC8647430 DOI: 10.1016/j.ajpath.2021.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022]
Abstract
B cell-activating factor (BAFF), part of a tumor necrosis factor family of cytokines, was recently identified as a regulator of atherosclerosis; however, its role in aortic aneurysm has not been determined. Here, the study examined the effect of selective BAFF antagonism using an anti-BAFF antibody (blocks binding of BAFF to receptors BAFF receptor 3, transmembrane activator and CAML interactor, and B-cell maturation antigen) and mBaffR-mFc (blocks binding of BAFF to BAFF receptor 3) on a murine model of abdominal aortic aneurysm (AAA). In a prevention strategy, the antagonists were injected before the induction of AAA, and in an intervention strategy, the antagonists were injected after the induction of AAA. Both strategies attenuated the formation of AAA. In the intervention group, BAFF antagonism depleted most of the mature B-cell subsets in spleen and circulation, leading to enhanced resolution of inflammation in AAA as indicated by decreased infiltration of B cells and proinflammatory macrophages and a reduced number of apoptotic cells. In AAA tissues, B cells and macrophages were found in close contact. In vitro, B cells, irrespective of treatment with BAFF, impaired the efferocytosis activity of macrophages, suggesting a direct innate role of B cells on macrophage function. Altogether, BAFF antagonism affects survival of the mature B cells, promotes resolution of inflammation in the aorta, and attenuates the growth of AAA in mice.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/therapy
- B-Cell Activating Factor/antagonists & inhibitors
- B-Cell Activating Factor/genetics
- B-Cell Activating Factor/immunology
- B-Cell Activating Factor/physiology
- B-Lymphocyte Subsets/pathology
- Cell Count
- Cells, Cultured
- Chemotaxis, Leukocyte/physiology
- Disease Models, Animal
- Disease Progression
- Humans
- Immunoglobulin Fc Fragments/pharmacology
- Immunoglobulin Fc Fragments/therapeutic use
- Macrophages/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
Collapse
Affiliation(s)
- Michael D Spinosa
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | | | - Melissa Lempicki
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina
| | - Prasad Srikakulapu
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Matthew J Johnsrude
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina
| | - Coleen A McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Gilbert R Upchurch
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Gorav Ailawadi
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Akshaya K Meher
- Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina; Department of Pharmacology, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
29
|
Lin J, Tang B, Feng Z, Hao W, Hu W. Decreased B lymphocytes subpopulations are associated with higher atherosclerotic risk in elderly patients with moderate-to-severe chronic kidney diseases. BMC Nephrol 2021; 22:396. [PMID: 34844574 PMCID: PMC8630907 DOI: 10.1186/s12882-021-02613-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/12/2021] [Indexed: 11/11/2022] Open
Abstract
Aim Cardiovascular diseases (CVD) are the leading cause of death in patients with chronic kidney disease (CKD), and the risk of CVD increases with reductions in renal function. This study aims to investigate the potential roles of B lymphocyte populations in subclinical atherosclerosis (measured by intima-media thickness, IMT) and prognosis in elderly patients with moderate-to-severe CKD. Methods In this study, a total of 219 patients (143 moderate-to-severe CKD patients with stage 3–4 and 76 non-CKD controls) were recruited. B cell subsets: CD19(+)CD5(+) and CD19(+)CD5(−) B cells were analyzed by flow cytometry. Intima-media thickness (IMT) was measured by ultrasound. Correlations between the B cell subsets with IMT and clinical outcome was analyzed. Results CKD patients showed increased IMT (P = 0.006). The level of CD19(+)CD5(+) and CD19(+)CD5(−) B cells were decreased in CKD patients. Correlation analysis showed that IMT was positively correlated with systolic blood pressure, protein/creatinine ratio and diabetes (P < 0.05), and were negatively correlated with CD19(+)CD5(+) and CD19(+)CD5(−) B lymphocytes (P < 0.05). Stepwise multiple regression analysis showed that CD19(+)CD5(−) B cells had a significant independent association with IMT (P < 0.05). IMT was increased in lower level of total CD19(+) B cells (≤ 0.06 × 109 /L) and CD19(+)CD5(−) B cells (≤ 0.05 × 109 /L) (P < 0.05). Kaplan-Meier analysis showed that patients with lower levels of CD19(+)CD5(+) and CD19(+)CD5(−) B cells exhibited worse survival (P < 0.05). Cox regression analysis showed that patients with lower CD19(+)CD5(+) and CD19(+)CD5(−) B cells counts have a higher risk of all-cause mortality (P < 0.05). Conclusions Our results showed that decreased CD19(+)CD5(+) and CD19(+)CD5(−) B lymphocytes were correlated with atherosclerosis and worse survival, which indicates that B lymphocytes might involve in atherosclerosis and associated the prognosis of elderly patients with moderate-to-severe CKD.
Collapse
Affiliation(s)
- Jieshan Lin
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, 510080, China.,Department of Nephrology, Blood Purification Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Bin Tang
- Department of Nephrology, Blood Purification Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Zhanwu Feng
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Wenke Hao
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, 510080, China
| | - Wenxue Hu
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, 510080, China.
| |
Collapse
|
30
|
de Mol J, Kuiper J, Tsiantoulas D, Foks AC. The Dynamics of B Cell Aging in Health and Disease. Front Immunol 2021; 12:733566. [PMID: 34675924 PMCID: PMC8524000 DOI: 10.3389/fimmu.2021.733566] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022] Open
Abstract
Aging is considered to be an important risk factor for several inflammatory diseases. B cells play a major role in chronic inflammatory diseases by antibody secretion, antigen presentation and T cell regulation. Different B cell subsets have been implicated in infections and multiple autoimmune diseases. Since aging decreases B cell numbers, affects B cell subsets and impairs antibody responses, the aged B cell is expected to have major impacts on the development and progression of these diseases. In this review, we summarize the role of B cells in health and disease settings, such as atherosclerotic disease. Furthermore, we provide an overview of age-related changes in B cell development and function with respect to their impact in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jill de Mol
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | | | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
31
|
Porsch F, Mallat Z, Binder CJ. Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies. Cardiovasc Res 2021; 117:2544-2562. [PMID: 34450620 DOI: 10.1093/cvr/cvab285] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune mechanisms are critically involved in the pathogenesis of atherosclerosis and its clinical manifestations. Associations of specific antibody levels and defined B cell subsets with cardiovascular disease activity in humans as well as mounting evidence from preclinical models demonstrate a role of B cells and humoral immunity in atherosclerotic cardiovascular disease. These include all aspects of B cell immunity, the generation of antigen-specific antibodies, antigen presentation and co-stimulation of T cells, as well as production of cytokines. Through their impact on adaptive and innate immune responses and the regulation of many other immune cells, B cells mediate both protective and detrimental effects in cardiovascular disease. Several antigens derived from (oxidised) lipoproteins, the vascular wall and classical autoantigens have been identified. The unique antibody responses they trigger and their relationship with atherosclerotic cardiovascular disease are reviewed. In particular, we focus on the different effector functions of specific IgM, IgG, and IgE antibodies and the cellular responses they trigger and highlight potential strategies to target B cell functions for therapy.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Unversité Paris Descartes, Sorbonne Paris Cité, Paris France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Abstract
B cells are traditionally known for their ability to produce antibodies in the context of adaptive immune responses. However, over the last decade B cells have been increasingly recognized as modulators of both adaptive and innate immune responses, as well as players in an important role in the pathogenesis of a variety of human diseases. Here, after briefly summarizing our current understanding of B cell biology, we present a systematic review of the literature from both animal models and human studies that highlight the important role that B lymphocytes play in cardiac and vascular disease. While many aspects of B cell biology in the vasculature and, to an even greater extent, in the heart remain unclear, B cells are emerging as key regulators of cardiovascular adaptation to injury.
Collapse
Affiliation(s)
- Luigi Adamo
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; , ,
| | - Cibele Rocha-Resende
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; , ,
| | - Douglas L Mann
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA; , ,
| |
Collapse
|
33
|
Deroissart J, Porsch F, Koller T, Binder CJ. Anti-inflammatory and Immunomodulatory Therapies in Atherosclerosis. Handb Exp Pharmacol 2021; 270:359-404. [PMID: 34251531 DOI: 10.1007/164_2021_505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypercholesterolemia is a major risk factor in atherosclerosis development and lipid-lowering drugs (i.e., statins) remain the treatment of choice. Despite effective reduction of LDL cholesterol in patients, a residual cardiovascular risk persists in some individuals, highlighting the need for further therapeutic intervention. Recently, the CANTOS trial paved the way toward the development of specific therapies targeting inflammation, a key feature in atherosclerosis progression. The pre-existence of multiple drugs modulating both innate and adaptive immune responses has significantly accelerated the number of translational studies applying these drugs to atherosclerosis. Additional preclinical research has led to the discovery of new therapeutic targets, offering promising perspectives for the treatment and prevention of atherosclerosis. Currently, both drugs with selective targeting and broad unspecific anti-inflammatory effects have been tested. In this chapter, we aim to give an overview of current advances in immunomodulatory treatment approaches for atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Koller
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
34
|
Pattarabanjird T, Li C, McNamara C. B Cells in Atherosclerosis: Mechanisms and Potential Clinical Applications. ACTA ACUST UNITED AC 2021; 6:546-563. [PMID: 34222726 PMCID: PMC8246059 DOI: 10.1016/j.jacbts.2021.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
Abstract
B cells regulate atherosclerotic plaque formation through production of antibodies and cytokines, and effects are subset specific (B1 and B2). Putative human atheroprotective B1 cells function similarly to murine B1 in their spontaneous IgM antibody production. However, marker strategies in identifying human and murine B1 are different. IgM antibody to oxidation specific epitopes produced by B1 cells associate with human coronary artery disease. Neoantigen immunization may be a promising strategy for atherosclerosis vaccine development, but further study to determine relevant antigens still need to be done. B-cell–targeted therapies, used in treating autoimmune diseases as well as lymphoid cancers, might have potential applications in treating cardiovascular diseases. Short- and long-term cardiovascular effects of these agents need to be assessed.
Because atherosclerotic cardiovascular disease is a leading cause of death worldwide, understanding inflammatory processes underpinning its pathology is critical. B cells have been implicated as a key immune cell type in regulating atherosclerosis. B-cell effects, mediated by antibodies and cytokines, are subset specific. In this review, we focus on elaborating mechanisms underlying subtype-specific roles of B cells in atherosclerosis and discuss available human data implicating B cells in atherosclerosis. We further discuss potential B cell–linked therapeutic approaches, including immunization and B cell–targeted biologics. Given recent evidence strongly supporting a role for B cells in human atherosclerosis and the expansion of immunomodulatory agents that affect B-cell biology in clinical use and clinical trials for other disorders, it is important that the cardiovascular field be cognizant of potential beneficial or untoward effects of modulating B-cell activity on atherosclerosis.
Collapse
Key Words
- APRIL, A proliferation−inducing ligand
- ApoE, apolipoprotein E
- B-cell
- BAFF, B-cell–activating factor
- BAFFR, B-cell–activating factor receptor
- BCMA, B-cell maturation antigen
- BCR, B-cell receptor
- Breg, regulatory B cell
- CAD, coronary artery disease
- CTLA4, cytotoxic T-lymphocyte–associated protein 4
- CVD, cardiovascular disease
- CXCR4, C-X-C motif chemokine receptor 4
- GC, germinal center
- GITR, glucocorticoid-induced tumor necrosis factor receptor–related protein
- GITRL, glucocorticoid-induced tumor necrosis factor receptor–related protein ligand
- GM-CSF, granulocyte-macrophage colony–stimulating factor
- ICI, immune checkpoint inhibitor
- IFN, interferon
- IL, interleukin
- IVUS, intravascular ultrasound
- LDL, low-density lipoprotein
- LDLR, low-density lipoprotein receptor
- MDA-LDL, malondialdehyde-modified low-density lipoprotein
- MI, myocardial infarction
- OSE, oxidation-specific epitope
- OxLDL, oxidized low-density lipoprotein
- PC, phosphorylcholine
- PD-1, programmed cell death protein 1
- PD-L2, programmed death ligand 2
- PDL1, programmed death ligand 1
- RA, rheumatoid arthritis
- SLE, systemic lupus erythematosus
- TACI, transmembrane activator and CAML interactor
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- atherosclerosis
- immunoglobulins
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Cynthia Li
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Coleen McNamara
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
35
|
Petkevicius K, Bidault G, Virtue S, Newland SA, Dale M, Dugourd A, Saez-Rodriguez J, Mallat Z, Vidal-Puig A. Macrophage beta2-adrenergic receptor is dispensable for the adipose tissue inflammation and function. Mol Metab 2021; 48:101220. [PMID: 33774223 PMCID: PMC8086137 DOI: 10.1016/j.molmet.2021.101220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Neuroimmune interactions between the sympathetic nervous system (SNS) and macrophages are required for the homeostasis of multiple tissues, including the adipose tissue. It has been proposed that the SNS maintains adipose tissue macrophages (ATMs) in an anti-inflammatory state via direct norepinephrine (NE) signaling to macrophages. This study aimed to investigate the physiological importance of this paradigm by utilizing a mouse model in which the adrenergic signaling from the SNS to macrophages, but not to other adipose tissue cells, was disrupted. METHODS We generated a macrophage-specific B2AR knockout mouse (Adrb2ΔLyz2) by crossing Adrb2fl/fl and Lyz2Cre/+ mice. We have previously shown that macrophages isolated from Adrb2ΔLyz2 animals do not respond to NE stimulation in vitro. Herein we performed a metabolic phenotyping of Adrb2ΔLyz2 mice on either chow or high-fat diet (HFD). We also assessed the adipose tissue function of Adrb2ΔLyz2 animals during fasting and cold exposure. Finally, we transplanted Adrb2ΔLyz2 bone marrow to low-density lipoprotein receptor (LDLR) knockout mice and investigated the development of atherosclerosis during Western diet feeding. RESULTS We demonstrated that SNS-associated ATMs have a transcriptional profile indicative of activated beta-2 adrenergic receptor (B2AR), the main adrenergic receptor isoform in myeloid cells. However, Adrb2ΔLyz2 mice have unaltered energy balance on a chow or HFD. Furthermore, Adrb2ΔLyz2 mice show similar levels of adipose tissue inflammation and function during feeding, fasting, or cold exposure, and develop insulin resistance during HFD at the same rate as controls. Finally, macrophage-specific B2AR deletion does not affect the development of atherosclerosis on an LDL receptor-null genetic background. CONCLUSIONS Overall, our data suggest that the SNS does not directly modulate the phenotype of adipose tissue macrophages in either lean mice or mouse models of cardiometabolic disease. Instead, sympathetic nerve activity exerts an indirect effect on adipose tissue macrophages through the modulation of adipocyte function.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adipose Tissue, White/metabolism
- Animals
- Atherosclerosis/complications
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Bone Marrow Transplantation/methods
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Diet, Western/adverse effects
- Disease Models, Animal
- Female
- Insulin Resistance/genetics
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/complications
- Obesity/genetics
- Obesity/metabolism
- Panniculitis/genetics
- Panniculitis/metabolism
- Phenotype
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/genetics
- Sympathetic Nervous System/metabolism
Collapse
Affiliation(s)
- Kasparas Petkevicius
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom.
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Sam Virtue
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Stephen A Newland
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin Dale
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Aurelien Dugourd
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine & Heidelberg University Hospital, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine & Heidelberg University Hospital, Heidelberg, Germany
| | - Ziad Mallat
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, United Kingdom.
| |
Collapse
|
36
|
Srikakulapu P, Upadhye A, Drago F, Perry HM, Bontha SV, McSkimming C, Marshall MA, Taylor AM, McNamara CA. Chemokine Receptor-6 Promotes B-1 Cell Trafficking to Perivascular Adipose Tissue, Local IgM Production and Atheroprotection. Front Immunol 2021; 12:636013. [PMID: 33679793 PMCID: PMC7933012 DOI: 10.3389/fimmu.2021.636013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Chemokine receptor-6 (CCR6) mediates immune cell recruitment to inflammatory sites and has cell type-specific effects on diet-induced atherosclerosis in mice. Previously we showed that loss of CCR6 in B cells resulted in loss of B cell-mediated atheroprotection, although the B cell subtype mediating this effect was unknown. Perivascular adipose tissue (PVAT) harbors high numbers of B cells including atheroprotective IgM secreting B-1 cells. Production of IgM antibodies is a major mechanism whereby B-1 cells limit atherosclerosis development. Yet whether CCR6 regulates B-1 cell number and production of IgM in the PVAT is unknown. In this present study, flow cytometry experiments demonstrated that both B-1 and B-2 cells express CCR6, albeit at a higher frequency in B-2 cells in both humans and mice. Nevertheless, B-2 cell numbers in peritoneal cavity (PerC), spleen, bone marrow and PVAT were no different in ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. In contrast, the numbers of atheroprotective IgM secreting B-1 cells were significantly lower in the PVAT of ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. Surprisingly, adoptive transfer (AT) of CD43- splenic B cells into B cell-deficient μMT -/- ApoE -/- mice repopulated the PerC with B-1 and B-2 cells and reduced atherosclerosis when transferred into ApoE -/- CCR6 +/+ sIgM -/- mice only when those cells expressed both CCR6 and sIgM. CCR6 expression on circulating human B cells in subjects with a high level of atherosclerosis in their coronary arteries was lower only in the putative human B-1 cells. These results provide evidence that B-1 cell CCR6 expression enhances B-1 cell number and IgM secretion in PVAT to provide atheroprotection in mice and suggest potential human relevance to our murine findings.
Collapse
Affiliation(s)
- Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Aditi Upadhye
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Heather M Perry
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Sai Vineela Bontha
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Chantel McSkimming
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Melissa A Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Angela M Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Coleen A McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
37
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|
38
|
Saidoune F, Even G, Lamri Y, Chezel J, Gaston AT, Escoubet B, Papo T, Charles N, Nicoletti A, Sacre K. Effects of BAFF Neutralization on Atherosclerosis Associated With Systemic Lupus Erythematosus. Arthritis Rheumatol 2020; 73:255-264. [PMID: 32783382 DOI: 10.1002/art.41485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/06/2020] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) is the leading cause of death in systemic lupus erythematosus (SLE). B cells play a key role in the pathogenesis of lupus, and anti-BAFF therapy has been approved for use in SLE. Since mature B cells also promote atherosclerosis, we undertook this study to evaluate, in a mouse model and in SLE patients, whether BAFF neutralization has an atheroprotective effect in SLE. METHODS The effect of BAFF on atherosclerosis associated with lupus was investigated in the atherosclerosis/lupus-prone apolipoprotein E-knockout D227K mouse model and in a cohort of SLE patients. Mice were treated with a blocking anti-BAFF monoclonal antibody (mAb), while fed a standard chow diet. Carotid plaque and carotid intima-media thickness were assessed by ultrasound at baseline and during follow-up in SLE patients who were asymptomatic for CVD. RESULTS Anti-BAFF mAb in ApoE-/- D227K mice induced B cell depletion, efficiently treated lupus, and improved atherosclerosis lesions (21% decrease; P = 0.007) in mice with low plasma cholesterol levels but worsened the lesions (17% increase; P = 0.06) in mice with high cholesterol levels. The atheroprotective effect of the BAFF-BAFF receptor signaling inhibition on B cells was counterbalanced by the proatherogenic effect of the BAFF-TACI signaling inhibition on macrophages. In SLE patients, blood BAFF levels were associated with subclinical atherosclerosis (r = 0.26, P = 0.03). Anti-BAFF mAb treatment had a differential effect on the intima-media thickness progression in SLE patients depending on body mass index. CONCLUSION Depending on the balance between lipid-induced and B cell-induced proatherogenic conditions, anti-BAFF could be detrimental or beneficial, respectively, to atherosclerosis development in SLE.
Collapse
Affiliation(s)
- Fanny Saidoune
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Guillaume Even
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Yasmine Lamri
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Julie Chezel
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Anh-Thu Gaston
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Brigitte Escoubet
- Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Thomas Papo
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Nicolas Charles
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Antonino Nicoletti
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Karim Sacre
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| |
Collapse
|
39
|
Nus M, Basatemur G, Galan M, Cros-Brunsó L, Zhao TX, Masters L, Harrison J, Figg N, Tsiantoulas D, Geissmann F, Binder CJ, Sage AP, Mallat Z. NR4A1 Deletion in Marginal Zone B Cells Exacerbates Atherosclerosis in Mice-Brief Report. Arterioscler Thromb Vasc Biol 2020; 40:2598-2604. [PMID: 32907369 PMCID: PMC7571845 DOI: 10.1161/atvbaha.120.314607] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. NR4A orphan receptors have been well studied in vascular and myeloid cells where they play important roles in the regulation of inflammation in atherosclerosis. NR4A1 (nerve growth factor IB) is among the most highly induced transcription factors in B cells following BCR (B-cell receptor) stimulation. Given that B cells substantially contribute to the development of atherosclerosis, we examined whether NR4A1 regulates B-cell function during atherogenesis.
Collapse
Affiliation(s)
- Meritxell Nus
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.).,CIBER de Enfermedades Cardiovasculares, Spain (M.N., M.G.)
| | - Gemma Basatemur
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Maria Galan
- CIBER de Enfermedades Cardiovasculares, Spain (M.N., M.G.).,Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (M.G.)
| | - Laia Cros-Brunsó
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Leanne Masters
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - James Harrison
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Nichola Figg
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Dimitrios Tsiantoulas
- Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., C.J.B.)
| | | | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria (D.T., C.J.B.)
| | - Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (M.N., G.B., L.C.-B., T.X.Z., L.M., J.H., N.F., A.P.S., Z.M.)
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a complex disease process with lipid as a traditional modifiable risk factor and therapeutic target in treating atherosclerotic cardiovascular disease (ACVD). Recent evidence indicates that genetic influence and host immune response also are vital in this process. How these elements interact and modify each other and if immune response may emerge as a novel modifiable target remain poorly understood. RECENT FINDINGS Numerous preclinical studies have clearly demonstrated that hypercholesterolemia is essential for atherogenesis, but genetic variations and host immune-inflammatory responses can modulate the pro-atherogenic effect of elevated LDL-C. Clinical studies also suggest that a similar paradigm may also be operational in atherogenesis in humans. More importantly each element modifies the biological behavior of the other two elements, forming a triangular relationship among the three. Modulating any one of them will have downstream impact on atherosclerosis. This brief review summarizes the relationship among lipids, genes, and immunity in atherogenesis and presents evidence to show how these elements affect each other. Modulation of immune response, though in its infancy, has a potential to emerge as a novel clinical strategy in treating ACVD.
Collapse
|
41
|
Mangge H, Prüller F, Schnedl W, Renner W, Almer G. Beyond Macrophages and T Cells: B Cells and Immunoglobulins Determine the Fate of the Atherosclerotic Plaque. Int J Mol Sci 2020; 21:ijms21114082. [PMID: 32521607 PMCID: PMC7312004 DOI: 10.3390/ijms21114082] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis (AS) leading to myocardial infarction and stroke remains worldwide the main cause for mortality. Vulnerable atherosclerotic plaques are responsible for these life-threatening clinical endpoints. Atherosclerosis is a chronic, complex, inflammatory disease with interactions between metabolic dysfunction, dyslipidemia, disturbed microbiome, infectious triggers, vascular, and immune cells. Undoubtedly, the immune response is a most important piece of the pathological puzzle in AS. Although macrophages and T cells have been the focus of research in recent years, B cells producing antibodies and regulating T and natural killer (NKT) cell activation are more important than formerly thought. New results show that the B cells exert a prominent role with atherogenic and protective facets mediated by distinct B cell subsets and different immunoglobulin effects. These new insights come, amongst others, from observations of the effects of innovative B cell targeted therapies in autoimmune diseases like systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). These diseases associate with AS, and the beneficial side effects of B cell subset depleting (modifying) therapies on atherosclerotic concomitant disease, have been observed. Moreover, the CANTOS study (NCT01327846) showed impressive results of immune-mediated inflammation as a new promising target of action for the fight against atherosclerotic endpoints. This review will reflect the putative role of B cells in AS in an attempt to connect observations from animal models with the small spectrum of the thus far available human data. We will also discuss the clinical therapeutic potency of B cell modulations on the process of AS.
Collapse
Affiliation(s)
- Harald Mangge
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (F.P.); (W.R.); (G.A.)
- Correspondence: ; Tel.: +43-664-3373531
| | - Florian Prüller
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (F.P.); (W.R.); (G.A.)
| | - Wolfgang Schnedl
- Department of Internal Medicine, Practice for General Internal Medicine, 8600 Bruck/Mur, Austria;
| | - Wilfried Renner
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (F.P.); (W.R.); (G.A.)
| | - Gunter Almer
- Clinical Institute for Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (F.P.); (W.R.); (G.A.)
| |
Collapse
|
42
|
Tay C, Kanellakis P, Hosseini H, Cao A, Toh BH, Bobik A, Kyaw T. B Cell and CD4 T Cell Interactions Promote Development of Atherosclerosis. Front Immunol 2020; 10:3046. [PMID: 31998318 PMCID: PMC6965321 DOI: 10.3389/fimmu.2019.03046] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
Interaction between B and CD4 T cells is crucial for their optimal responses in adaptive immunity. Immune responses augmented by their partnership promote chronic inflammation. Here we report that interaction between B and CD4 T cells augments their atherogenicity to promote lipid-induced atherosclerosis. Genetic deletion of the gene encoding immunoglobulin mu (μ) heavy chain (μMT) in ApoE−/− mice resulted in global loss of B cells including those in atherosclerotic plaques, undetectable immunoglobulins and impaired germinal center formation. Despite unaffected numbers in the circulation and peripheral lymph nodes, CD4 T cells were also reduced in spleens as were activated and memory CD4 T cells. In hyperlipidemic μMT−/− ApoE−/− mice, B cell deficiency decreased atherosclerotic lesions, accompanied by absence of immunoglobulins and reduced CD4 T cell accumulation in lesions. Adoptive transfer of B cells deficient in either MHCII or co-stimulatory molecule CD40, molecules required for B and CD4 T cell interaction, into B cell-deficient μMT−/− ApoE−/− mice failed to increase atherosclerosis. In contrast, wildtype B cells transferred into μMT−/− ApoE−/− mice increased atherosclerosis and increased CD4 T cells in lesions including activated and memory CD4 T cells. Transferred B cells also increased their expression of atherogenic cytokines IL-1β, TGF-β, MCP-1, M-CSF, and MIF, with partial restoration of germinal centers and plasma immunoglobulins. Our study demonstrates that interaction between B and CD4 T cells utilizing MHCII and CD40 is essential to augment their function to increase atherosclerosis in hyperlipidemic mice. These findings suggest that targeting B cell and CD4 T cell interaction may be a therapeutic strategy to limit atherosclerosis progression.
Collapse
Affiliation(s)
- Christopher Tay
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hamid Hosseini
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Ramel D, Gayral S, Sarthou MK, Augé N, Nègre-Salvayre A, Laffargue M. Immune and Smooth Muscle Cells Interactions in Atherosclerosis: How to Target a Breaking Bad Dialogue? Front Pharmacol 2019; 10:1276. [PMID: 31824304 PMCID: PMC6882774 DOI: 10.3389/fphar.2019.01276] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a well-known pathophysiological factor of atherosclerosis but its therapeutic targeting has long been ignored. However, recent advances in the understanding of the immune mechanisms implicated in atherosclerosis have unveiled several therapeutic targets currently undergoing clinical trials. These studies have also shed light on a dialogue between the immune compartment and vascular smooth muscle cells (VSMCs) that plays a critical role in atherosclerotic disease initiation, progression, and stabilization. Our review focuses on the link between cellular and soluble immune effectors and VSMC behavior at different phases of the pathology. Furthermore, we discuss the potential targeting of these interactions to efficiently prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Damien Ramel
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Stéphanie Gayral
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Marie-Kerguelen Sarthou
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Nathalie Augé
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Anne Nègre-Salvayre
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Muriel Laffargue
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| |
Collapse
|
44
|
Spinelli FR, Barbati C, Cecarelli F, Morello F, Colasanti T, Vomero M, Massaro L, Orefice V, Alessandri C, Valesini G, Conti F. B lymphocyte stimulator modulates number and function of endothelial progenitor cells in systemic lupus erythematosus. Arthritis Res Ther 2019; 21:245. [PMID: 31752963 PMCID: PMC6868730 DOI: 10.1186/s13075-019-2015-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/24/2019] [Indexed: 01/10/2023] Open
Abstract
Background Circulating endothelial progenitor cells (EPCs) are biologic markers of endothelial function. In patients with systemic lupus erythematosus (SLE), the numerical reduction and functional impairment of EPCs contribute to the endothelial dysfunction. Through ex vivo and in vitro studies, we aimed at evaluating the effects of B lymphocyte stimulator (BLyS) on EPC colonies and endothelial cells and also investigating BLyS receptor expression on these cells. Methods EPCs were isolated from peripheral blood mononuclear cells (PBMC). In order to evaluate their ability to form colonies, EPCs were cultured on fibronectin-coated dishes and incubated with BlyS alone or BlyS and belimumab. Apoptosis of EPCs and endothelial cell line EA.hy926 was evaluated after 6, 12, and 24 h of incubation with BLyS and after 6 h with BLyS and belimumab. The expression of B cell activating factor-receptor (BAFF-R), B cell maturation antigen (BCMA), and transmembrane activator and calcium modulator and cyclophilin ligand (CAML) interactor (TACI) on EPCs and EA.hy926 was analyzed by cytofluorimetry. Results The number of EPC colonies was lower in patients than in controls. Moreover, the colonies from SLE patients were poorly organized compared to controls; the addition of belimumab restored the colony structure. Incubation with BLyS induced apoptosis of EPCs and EA.hy926 that was inhibited by the co-incubation with belimumab. BAFF-R and BCMA were expressed on both EPCs and EA.hy926, while TACI was expressed only on EPCs. Conclusions EPCs and endothelial cells preferentially express BAFF-R which could be involved in the pro-apoptotic effect of BlyS. Belimumab administration seems to restore the quantitative and qualitative changes of EPC colonies both ex vivo and in vitro.
Collapse
Affiliation(s)
- Francesca Romana Spinelli
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy.
| | - Cristiana Barbati
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Fulvia Cecarelli
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Francesca Morello
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Tania Colasanti
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Marta Vomero
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Laura Massaro
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Valeria Orefice
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Department of Internal Medicine and Medical Specialties, Rheumatology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
45
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
46
|
Porsch F, Binder CJ. Impact of B-Cell–Targeted Therapies on Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:1705-1714. [DOI: 10.1161/atvbaha.119.311996] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by many immune cell subsets, including B cells. Therefore, targeting the inflammatory component of cardiovascular disease represents a promising therapeutic strategy. In the past years, immunotherapy has revolutionized the treatment of autoimmunity and cancer. Many of these clinically used strategies target B cells. Given the multifaceted role of B cells in atherogenesis, it is conceivable that B-cell–directed therapies can modulate disease development. Here, we review clinically available B-cell–targeted therapies and the possible benefits or detrimental effects on cardiovascular disease.
Collapse
Affiliation(s)
- Florentina Porsch
- From the Department for Laboratory Medicine, Medical University of Vienna, Austria (F.P., C.J.B.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (F.P., C.J.B.)
| | - Christoph J. Binder
- From the Department for Laboratory Medicine, Medical University of Vienna, Austria (F.P., C.J.B.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (F.P., C.J.B.)
| |
Collapse
|
47
|
Abdolmaleki F, Gheibi Hayat SM, Bianconi V, Johnston TP, Sahebkar A. Atherosclerosis and immunity: A perspective. Trends Cardiovasc Med 2019; 29:363-371. [DOI: 10.1016/j.tcm.2018.09.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/09/2018] [Accepted: 09/25/2018] [Indexed: 01/11/2023]
|
48
|
Bagchi-Chakraborty J, Francis A, Bray T, Masters L, Tsiantoulas D, Nus M, Harrison J, Broekhuizen M, Leggat J, Clatworthy MR, Espéli M, Smith KG, Binder CJ, Mallat Z, Sage AP. B Cell Fcγ Receptor IIb Modulates Atherosclerosis in Male and Female Mice by Controlling Adaptive Germinal Center and Innate B-1-Cell Responses. Arterioscler Thromb Vasc Biol 2019; 39:1379-1389. [PMID: 31092015 PMCID: PMC6636804 DOI: 10.1161/atvbaha.118.312272] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/22/2019] [Indexed: 02/02/2023]
Abstract
Objective- Investigate the impact of modulating B cell FcγRIIb (Fcγ receptor IIb) expression on atherosclerosis. Approach and Results- Western diet-induced atherosclerosis was assessed in Ldlr-/- or Apoe-/- mice with B cell-specific overexpression of FcγRIIb or with an FcγRIIb promoter mutation that alters FcγRIIb expression in germinal center (GC) B cells. In males, overexpression of FcγRIIb on B cells severely reduced activated, class switched B cell responses, as indicated by reductions in GC B cells, plasma cells, and serum IgG but not IgM antibodies. Male mice overexpressing FcγRIIb developed less atherosclerosis, suggesting a pathogenic role for GC B cell IgG responses. In support of this hypothesis, male mice with a promoter polymorphism-driven reduction in FcγRIIb on GC B cells but not plasma cells have a converse phenotype of enhanced GC responses and IgG2c antibodies and enhanced atherosclerosis. IgG2c significantly enhanced TNF (tumor necrosis factor) secretion by CD11b+ CD11c+ cells expressing the high-affinity receptor FcγRIV. In females, overexpression of FcγRIIb on B cells not only reduced GC B cell responses but also substantially reduced B-1 cells and IgM antibodies, which translated into acceleration of atherosclerosis. Promoter-driven reduction in FcγRIIb did not alter GC B cell responses in females and, therefore, had no impact on atherosclerosis. Conclusions- B cell FcγRIIb differentially alters proatherogenic adaptive GC B cell and atheroprotective innate B-1 responses in male and female mice fed a western diet. Our results highlight the importance of a better understanding and ability to selectively target B cell responses in future immunotherapeutic approaches against human cardiovascular disease. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Jayashree Bagchi-Chakraborty
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Anna Francis
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Toni Bray
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Leanne Masters
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - James Harrison
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Michelle Broekhuizen
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Jennifer Leggat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Menna R. Clatworthy
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Marion Espéli
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France (M.E.)
| | - Kenneth G.C. Smith
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria (C.J.B.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (C.J.B.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
- Institut National de la Santé et de la Recherche Médicale, Universite Paris-Descartes, Paris Cardiovascular Research Center, and Université Paris-Descartes, France (Z.M.)
| | - Andrew P. Sage
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
49
|
Koulouri V, Koutsilieris M, Mavragani CP. B cells and atherosclerosis in systemic lupus erythematosus. Expert Rev Clin Immunol 2019; 15:417-429. [DOI: 10.1080/1744666x.2019.1571411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Vasiliki Koulouri
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, 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
| |
Collapse
|
50
|
Williams JW, Elvington A, Kessler S, Wohltmann M, Wu GF, Randolph GJ. B Cell-Mediated Antigen Presentation through MHC Class II Is Dispensable for Atherosclerosis Progression. Immunohorizons 2019; 3:37-44. [PMID: 31356175 PMCID: PMC6999615 DOI: 10.4049/immunohorizons.1800015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Depletion of B cells attenuates plaque development and modulates T cell responses in mouse models of atherosclerosis, suggesting that Ag presentation by B cells may promote disease progression. Thus, we set out to determine the role of B cell-mediated MHC class II (MHC II) Ag presentation during atherosclerotic plaque development. We developed murine conditional MHC II deletion and expression systems under control of the B cell-restricted CD19 promoter in an experimental model of atherosclerosis. Mice lacking MHC II expression only on B cells exhibited systemic shifts in germinal center and marginal zone B cell populations, leading to a reduced Ab response compared with littermate control animals. However, all populations were present and normal cholesterol uptake was detected in the plasma following high-fat diet treatment. In a second model, in which conditional expression of MHC II is limited only to B cells, showed similar overall cellularity characteristics compared with mice with complete MHC II deficiency. High-fat diet feeding showed no major changes in atherosclerotic plaque size or plaque cellular content in either conditional deletion or conditional expression approaches, compared with control animals. By testing the necessity and sufficiency of MHC II on B cells in the progression of atherosclerosis, we determine that MHC II on B cells does not directly regulate lesion development in murine models.
Collapse
Affiliation(s)
- Jesse W Williams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Skyler Kessler
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Mary Wohltmann
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Gregory F Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| |
Collapse
|