1
|
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
|
2
|
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 2024:10.1007/s11010-024-05158-y. [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] [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
|
3
|
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
|
4
|
Deroissart J, Binder CJ, Porsch F. Role of Antibodies and Their Specificities in Atherosclerotic Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2024; 44:2154-2168. [PMID: 39114917 DOI: 10.1161/atvbaha.124.319843] [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: 09/27/2024]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by innate and adaptive immunity including humoral immunity. Importantly, antibody alterations achieved by genetic means or active and passive immunization strategies in preclinical studies can improve or aggravate atherosclerosis. Additionally, a wide range of epidemiological data demonstrate not only an association between the total levels of different antibody isotypes but also levels of antibodies targeting specific antigens with atherosclerotic cardiovascular disease. Here, we discuss the potential role of atherogenic dyslipidemia on the antibody repertoire and review potential antibody-mediated effector mechanisms involved in atherosclerosis development highlighting the major atherosclerosis-associated antigens that trigger antibody responses.
Collapse
Affiliation(s)
- Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| |
Collapse
|
5
|
Kerns S, Owen KA, Daamen A, Kain J, Grammer AC, Lipsky PE. Genetic association with autoimmune diseases identifies molecular mechanisms of coronary artery disease. iScience 2024; 27:110715. [PMID: 39262791 PMCID: PMC11387803 DOI: 10.1016/j.isci.2024.110715] [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: 06/04/2024] [Revised: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Autoimmune patients have a significantly increased risk of developing coronary artery disease (CAD) compared to the general population. However, autoimmune patients often lack traditional risk factors for CAD and there is increasing recognition of inflammation in CAD development. In this study, we leveraged genome-wide association study (GWAS) data to understand whether there is a genetic relationship between CAD and autoimmunity. Statistical genetic comparison methods were used to identify correlated and causal SNPs between various autoimmune diseases and CAD. Pleiotropic SNPs were identified by cross-phenotype association analysis (CPASSOC) and overlap between GWAS. Causal SNPs were identified using Mendelian Randomization (MR) and Colocalization (COLOC). Using SNP-to-gene mapping, we additionally identified pleiotropic and causal genes and pathways associated between autoimmunity and CAD, which were contextualized by documentation of enrichment in individual cell types identified from coronary atherosclerotic plaques by single-cell RNA sequencing. These results provide insight into potential inflammatory therapeutic targets for CAD.
Collapse
Affiliation(s)
- Sophia Kerns
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Katherine A Owen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Andrea Daamen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Jessica Kain
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
- Stanford University Department of Genetics, Stanford, CA 94305, USA
| | - Amrie C Grammer
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Peter E Lipsky
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| |
Collapse
|
6
|
Chen Z, Wang Z, Cui Y, Xie H, Yi L, Zhu Z, Ni J, Du R, Wang X, Zhu J, Ding F, Quan W, Zhang R, Wang Y, Yan X. Serum BAFF level is associated with the presence and severity of coronary artery disease and acute myocardial infarction. BMC Cardiovasc Disord 2024; 24:471. [PMID: 39227771 PMCID: PMC11370111 DOI: 10.1186/s12872-024-04146-w] [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: 06/12/2023] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between circulating levels of B cell activating factor (BAFF) and the presence and severity of coronary artery disease (CAD) and acute myocardial infarction (AMI) in humans, as its biological functions in this context remain unclear. METHODS Serum BAFF levels were measured in a cohort of 723 patients undergoing angiography, including 204 patients without CAD (control group), 220 patients with stable CAD (CAD group), and 299 patients with AMI (AMI group). Logistic regression analyses were used to assess the association between BAFF and CAD or AMI. RESULTS Significantly elevated levels of BAFF were observed in patients with CAD and AMI compared to the control group. Furthermore, BAFF levels exhibited a positive correlation with the SYNTAX score (r = 0.3002, P < 0.0001) and the GRACE score (r = 0.5684, P < 0.0001). Logistic regression analysis demonstrated that increased BAFF levels were an independent risk factor for CAD (adjusted OR 1.305, 95% CI 1.078-1.580) and AMI (adjusted OR 2.874, 95% CI 1.708-4.838) after adjusting for confounding variables. Additionally, elevated BAFF levels were significantly associated with a high GRACE score (GRACE score 155 to 319, adjusted OR 4.297, 95% CI 1.841-10.030). BAFF exhibited a sensitivity of 75.0% and specificity of 71.4% in differentiating CAD patients with a high SYNTAX score, and a sensitivity of 75.5% and specificity of 72.8% in identifying AMI patients with a high GRACE score. CONCLUSION Circulating BAFF levels serve as a valuable diagnostic marker for CAD and AMI. Elevated BAFF levels are associated with the presence and severity of these conditions, suggesting its potential as a clinically relevant biomarker in cardiovascular disease.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ziyang Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuke Cui
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongyang Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Lei Yi
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jingwei Ni
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Run Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Weiwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
| | - Yueying Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
7
|
Ransegnola BP, Pattarabanjird T, McNamara CA. Tipping the Scale: Atheroprotective IgM-Producing B Cells in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1906-1915. [PMID: 39022832 PMCID: PMC11338718 DOI: 10.1161/atvbaha.124.319847] [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] [Indexed: 07/20/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease whose progression is fueled by proinflammatory moieties and limited by anti-inflammatory mediators. Whereas oxidative damage and the generation of oxidation-specific epitopes that act as damage-associated molecular patterns are highly inflammatory, IgM antibodies produced by B-1 and marginal zone B cells counteract unrestricted inflammation by neutralizing and encouraging clearance of these proinflammatory signals. In this review, we focus on describing the identities of IgM-producing B cells in both mice and humans, elaborating the mechanisms underlying IgM production, and discussing the potential strategies to augment the production of atheroprotective IgM. In addition, we will discuss promising therapeutic interventions in humans to help tip the scale toward augmentation of IgM production and to provide atheroprotection.
Collapse
Affiliation(s)
- Brett Patrick Ransegnola
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyaporn Pattarabanjird
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Coleen A. McNamara
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
8
|
Liu T, Chen Y, Hou L, Yu Y, Ma D, Jiang T, Zhao G. Immune cell-mediated features of atherosclerosis. Front Cardiovasc Med 2024; 11:1450737. [PMID: 39234608 PMCID: PMC11371689 DOI: 10.3389/fcvm.2024.1450737] [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: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by innate and adaptive immune responses, which seriously threatens human life and health. It is a primary cause of coronary heart disease, myocardial infarction, and peripheral vascular disease. Research has demonstrated that immune cells are fundamental to the development of atherosclerosis and chronic inflammation. Therefore, it is anticipated that immunotherapy targeting immune cells will be a novel technique in the management of atherosclerosis. This article reviews the growth of research on the regulatory role of immune cells in atherosclerosis and targeted therapy approaches. The purpose is to offer new therapeutic approaches for the control and treatment of cardiovascular illnesses caused by atherosclerosis.
Collapse
Affiliation(s)
- Tingting Liu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yanjun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lianjie Hou
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yulu Yu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
9
|
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
|
10
|
Jones PW, Mallat Z, Nus M. T-Cell/B-Cell Interactions in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1502-1511. [PMID: 38813700 PMCID: PMC11208060 DOI: 10.1161/atvbaha.124.319845] [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
Atherosclerosis is a complex inflammatory disease in which the adaptive immune response plays an important role. While the overall impact of T and B cells in atherosclerosis is relatively well established, we are only beginning to understand how bidirectional T-cell/B-cell interactions can exert prominent atheroprotective and proatherogenic functions. In this review, we will focus on these T-cell/B-cell interactions and how we could use them to therapeutically target the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Peter William Jones
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| | - Ziad Mallat
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
- INSERM U970, Paris Cardiovascular Research Centre, France (Z.M.)
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| |
Collapse
|
11
|
Xu X, Qiu F, Yang M, Liu X, Tao S, Zheng B. Unveiling Atherosclerotic Plaque Heterogeneity and SPP1 +/VCAN + Macrophage Subtype Prognostic Significance Through Integrative Single-Cell and Bulk-Seq Analysis. J Inflamm Res 2024; 17:2399-2426. [PMID: 38681071 PMCID: PMC11055562 DOI: 10.2147/jir.s454505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Background Dysregulated macrophages are important causes of Atherosclerosis (AS) formation and increased plaque instability, but the heterogeneity of these plaques and the role of macrophage subtypes in plaque instability have yet to be clarified. Methods This study integrates single-cell and bulk-seq data to analyze atherosclerotic plaques. Unsupervised clustering was used to reveal distinct plaque subtypes, while survival analysis and gene set variation analysis (GSVA) methods helped in understanding their clinical outcomes. Enrichment of differential expression of macrophage genes (DEMGs) score and pseudo-trajectory analysis were utilized to explore the biological functions and differentiation stages of macrophage subtypes in AS progression. Additionally, CellChat and the BayesPrism deconvolution method were used to elucidate macrophage subtype interaction and their prognostic significance at single-cell resolution. Finally, the expression of biomarkers was validated in mouse experiments. Results Three distinct AS plaque subtypes were identified, with cluster 3 plaque subtype being particularly associated with higher immune infiltration and poorer prognosis. The DEMGs score exhibited a significant elevation in three macrophage subtypes (SPP1+/VCAN+ macrophages, IL1B+ macrophages, and FLT3LG+ macrophages), associated with cluster 3 plaque subtype and highlighted the prognostic significance of these subtypes. Activation trajectory of the macrophage subtypes is divided into three states (Pre-branch, Cell fate 1, and Cell fate 2), and Cell fate 2 (SPP1+/VCAN+ macrophages, IL1B+ macrophages, and FLT3LG+ macrophages dominant) exhibiting the highest DEMGs score, distinct interactions with other cell components, and relating to poorer prognosis of ischemic events. This study also uncovered a unique SPP1+/VCAN+ macrophage subtype, rare in quantity but significant in influencing AS progression. Machine learning algorithms identified 10 biomarkers crucial for AS diagnosis. The validation of these biomarkers was performed using Mendelian Randomization analysis and in vitro methods, supporting their relevance in AS pathology. Conclusion Our study provides a comprehensive view of AS plaque heterogeneity and the prognostic significance of macrophage subtypes in plaque instability.
Collapse
Affiliation(s)
- Xiang Xu
- School of Medicine, Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Fuling Qiu
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Man Yang
- School of Medicine, Dali University, Dali City, Yunnan Province, People’s Republic of China
| | - Xiaoyong Liu
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Siming Tao
- Department of Cardiology, The Affiliated Hospital of Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
| | - Bingrong Zheng
- School of Medicine, Yunnan University, Kunming City, Yunnan Province, People’s Republic of China
| |
Collapse
|
12
|
Harrison J, Newland SA, Jiang W, Giakomidi D, Zhao X, Clement M, Masters L, Corovic A, Zhang X, Drago F, Ma M, Ozsvar Kozma M, Yasin F, Saady Y, Kothari H, Zhao TX, Shi GP, McNamara CA, Binder CJ, Sage AP, Tarkin JM, Mallat Z, Nus M. Marginal zone B cells produce 'natural' atheroprotective IgM antibodies in a T cell-dependent manner. Cardiovasc Res 2024; 120:318-328. [PMID: 38381113 PMCID: PMC10939463 DOI: 10.1093/cvr/cvae027] [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] [Received: 09/06/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The adaptive immune response plays an important role in atherosclerosis. In response to a high-fat/high-cholesterol (HF/HC) diet, marginal zone B (MZB) cells activate an atheroprotective programme by regulating the differentiation and accumulation of 'poorly differentiated' T follicular helper (Tfh) cells. On the other hand, Tfh cells activate the germinal centre response, which promotes atherosclerosis through the production of class-switched high-affinity antibodies. We therefore investigated the direct role of Tfh cells and the role of IL18 in Tfh differentiation in atherosclerosis. METHODS AND RESULTS We generated atherosclerotic mouse models with selective genetic deletion of Tfh cells, MZB cells, or IL18 signalling in Tfh cells. Surprisingly, mice lacking Tfh cells had increased atherosclerosis. Lack of Tfh not only reduced class-switched IgG antibodies against oxidation-specific epitopes (OSEs) but also reduced atheroprotective natural IgM-type anti-phosphorylcholine (PC) antibodies, despite no alteration of natural B1 cells. Moreover, the absence of Tfh cells was associated with an accumulation of MZB cells with substantially reduced ability to secrete antibodies. In the same manner, MZB cell deficiency in Ldlr-/- mice was associated with a significant decrease in atheroprotective IgM antibodies, including natural anti-PC IgM antibodies. In humans, we found a positive correlation between circulating MZB-like cells and anti-OSE IgM antibodies. Finally, we identified an important role for IL18 signalling in HF/HC diet-induced Tfh. CONCLUSION Our findings reveal a previously unsuspected role of MZB cells in regulating atheroprotective 'natural' IgM antibody production in a Tfh-dependent manner, which could have important pathophysiological and therapeutic implications.
Collapse
Affiliation(s)
- James Harrison
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Stephen A Newland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Wei Jiang
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Despoina Giakomidi
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xiaohui Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Clement
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Laboratory for Vascular Translational Sciences (LVTS), Université de Paris, INSERM U1148, Paris, France
| | - Leanne Masters
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrej Corovic
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xian Zhang
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrizio Drago
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Marcella Ma
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, UK
| | - Maria Ozsvar Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Froher Yasin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Yuta Saady
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hema Kothari
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tian X Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Coleen A McNamara
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andrew P Sage
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason M Tarkin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ziad Mallat
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- PARCC Inserm U970, Universite de Paris, Paris, France
| | - Meritxell Nus
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
13
|
Di Nunzio G, Hellberg S, Zhang Y, Ahmed O, Wang J, Zhang X, Björck HM, Chizh V, Schipper R, Aulin H, Francis R, Fagerberg L, Gisterå A, Metso J, Manfé V, Franco-Cereceda A, Eriksson P, Jauhiainen M, Hagberg CE, Olofsson PS, Malin SG. Kupffer cells dictate hepatic responses to the atherogenic dyslipidemic insult. NATURE CARDIOVASCULAR RESEARCH 2024; 3:356-371. [PMID: 39196121 PMCID: PMC11358021 DOI: 10.1038/s44161-024-00448-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/05/2024] [Indexed: 08/29/2024]
Abstract
Apolipoprotein-B (APOB)-containing lipoproteins cause atherosclerosis. Whether the vasculature is the initially responding site or if atherogenic dyslipidemia affects other organs simultaneously is unknown. Here we show that the liver responds to a dyslipidemic insult based on inducible models of familial hypercholesterolemia and APOB tracing. An acute transition to atherogenic APOB lipoprotein levels resulted in uptake by Kupffer cells and rapid accumulation of triglycerides and cholesterol in the liver. Bulk and single-cell RNA sequencing revealed a Kupffer-cell-specific transcriptional program that was not activated by a high-fat diet alone or detected in standard liver function or pathological assays, even in the presence of fulminant atherosclerosis. Depletion of Kupffer cells altered the dynamic of plasma and liver lipid concentrations, indicating that these liver macrophages help restrain and buffer atherogenic lipoproteins while simultaneously secreting atherosclerosis-modulating factors into plasma. Our results place Kupffer cells as key sentinels in organizing systemic responses to lipoproteins at the initiation of atherosclerosis.
Collapse
Affiliation(s)
- Giada Di Nunzio
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sanna Hellberg
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yuyang Zhang
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Osman Ahmed
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry, Faculty of Medicine, Khartoum University, Khartoum, Sudan
| | - Jiawen Wang
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xueming Zhang
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hanna M Björck
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Veronika Chizh
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ruby Schipper
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Aulin
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Roy Francis
- Science for Life Laboratory, Department of Cell and Molecular Biology (ICM), National Bioinformatics Infrastructure Sweden (NBIS), Uppsala University, Uppsala, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jari Metso
- Finnish Institute for Health and Welfare, Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | - Anders Franco-Cereceda
- Section of Cardiothoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matti Jauhiainen
- Finnish Institute for Health and Welfare, Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Carolina E Hagberg
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peder S Olofsson
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen G Malin
- Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Li H, Zhao Q, Liu D, Zhou B, Liao F, Chen L. Cathepsin B aggravates atherosclerosis in ApoE-deficient mice by modulating vascular smooth muscle cell pyroptosis through NF-κB / NLRP3 signaling pathway. PLoS One 2024; 19:e0294514. [PMID: 38165884 PMCID: PMC10760722 DOI: 10.1371/journal.pone.0294514] [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: 06/10/2023] [Accepted: 11/02/2023] [Indexed: 01/04/2024] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease involving cell death and inflammatory responses. Pyroptosis, a newly discovered pro-inflammatory programmed cell death process, exacerbates inflammatory responses. However, the roles of cathepsin B (CTSB) in pyroptosis and AS remain unclear. To gain further insight, we fed ApoE-/- mice a high-fat diet to investigate the effects and mechanisms of CTSB overexpression and silencing on AS. We also explored the specific role of CTSB in vascular smooth muscle cells (VSMCs) in vitro. The study revealed that high-fat diet led to the formation of AS plaques, and CTSB was found to increase the AS plaque lesion area. Immunohistochemical and TUNEL/caspase-1 staining revealed the existence of pyroptosis in atherosclerotic plaques, particularly in VSMCs. In vitro studies, including Hoechst 33342/propidium iodide staining, a lactate dehydrogenase (LDH) release assay, detection of protein indicators of pyroptosis, and detection of interleukin-1β (IL-1β) in cell culture medium, demonstrated that oxidized low-density lipoprotein (ox-LDL) induced VSMC pyroptosis. Additionally, CTSB promoted VSMC pyroptosis. Ox-LDL increased the expression of CTSB, which in turn activated the NOD-like receptor protein 3 (NLRP3) inflammasome and promoted NLRP3 expression by facilitating nuclear factor kappa B (NF-κB) p65 nuclear translocation. This effect could be attenuated by the NF-κB inhibitor SN50. Our research found that CTSB not only promotes VSMC pyroptosis by activating the NLRP3 inflammasome, but also increases the expression of NLRP3.
Collapse
Affiliation(s)
- Hui Li
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Quanwei Zhao
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Danan Liu
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Bo Zhou
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fujun Liao
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Long Chen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Institute of Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 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
|
Cheng XW, Narisawa M, Wang H, Piao L. Overview of multifunctional cysteinyl cathepsins in atherosclerosis-based cardiovascular disease: from insights into molecular functions to clinical implications. Cell Biosci 2023; 13:91. [PMID: 37202785 DOI: 10.1186/s13578-023-01040-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Cysteinyl cathepsins (CTSs) are widely known to have a proteolysis function that mediates recycling of unwanted proteins in endosomes and lysosomes, and investigation of CTSs has greatly improved with advances in live-imaging techniques both in vivo and in vitro, leading to three key findings. (1) CTSs are relocated from the lysosomes to other cellular spaces (i.e., cytosol, nucleus, nuclear membrane, plasma membrane, and extracellular milieu). (2) In addition to acidic cellular compartments, CTSs also exert biological activity in neutral environments. (3) CTSs also exert multiple nontraditional functions in, for example, extracellular matrix metabolism, cell signaling transduction, protein processing/trafficking, and cellular events. Various stimuli regulate the expression and activities of CTSs in vivo and vitro-e.g., inflammatory cytokines, oxidative stress, neurohormones, and growth factors. Accumulating evidence has confirmed the participation of CTSs in vascular diseases characterized by atherosclerosis, plaque rupture, thrombosis, calcification, aneurysm, restenosis/in-stent-restenosis, and neovasel formation. Circulating and tissue CTSs are promising as biomarkers and as a diagnostic imaging tool in patients with atherosclerosis-based cardiovascular disease (ACVD), and pharmacological interventions with their specific and non-specific inhibitors, and cardiovascular drugs might have potential for the therapeutic targeting of CTSs in animals. This review focuses on the update findings on CTS biology and the involvement of CTSs in the initiation and progression of ACVD and discusses the potential use of CTSs as biomarkers and small-molecule targets to prevent deleterious nontraditional functions in ACVD.
Collapse
Affiliation(s)
- Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China.
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, Jilin PR. 133000, China.
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4668550, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| |
Collapse
|
17
|
Meng Q, Liu H, Liu J, Pang Y, Liu Q. Advances in immunotherapy modalities for atherosclerosis. Front Pharmacol 2023; 13:1079185. [PMID: 36703734 PMCID: PMC9871313 DOI: 10.3389/fphar.2022.1079185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Atherosclerosis is the pathological basis of atherosclerotic cardiovascular disease (ASCVD). Atherosclerosis is now understood to be a long-term immune-mediated inflammatory condition brought on by a complicated chain of factors, including endothelial dysfunction, lipid deposits in the artery wall, and monocyte-derived macrophage infiltration, in which both innate immunity and adaptive immunity play an indispensable role. Recent studies have shown that atherosclerosis can be alleviated by inducing a protective immune response through certain auto-antigens or exogenous antigens. Some clinical trials have also demonstrated that atherosclerotic is associated with the presence of immune cells and immune factors in the body. Therefore, immunotherapy is expected to be a new preventive and curative measure for atherosclerosis. In this review, we provide a summary overview of recent progress in the research of immune mechanisms of atherosclerosis and targeted therapeutic pathways.
Collapse
Affiliation(s)
- Qingwen Meng
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China,Deparment of Cardiovascular, The First Affiliated Hospital of Hainan Medical University, Haikou, China,Hainan Provincial Key Laboratory of Tropical Brain Research and Transformation, Hainan Medical University, Haikou, China
| | - Huajiang Liu
- Deparment of Cardiovascular, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jinteng Liu
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Yangyang Pang
- School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China,School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China,*Correspondence: Qibing Liu,
| |
Collapse
|
18
|
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: 7] [Impact Index Per Article: 2.3] [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
|
19
|
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
|
20
|
Tian C, Mahara G, Zhang H, Tan X. Association of immunoglobulin G N-glycosylation with carotid atherosclerotic plaque phenotypes and actual clinical cardiovascular events: a study protocol for a longitudinal prospective cohort study. BMJ Open 2022; 12:e058922. [PMID: 35868824 PMCID: PMC9316026 DOI: 10.1136/bmjopen-2021-058922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 07/01/2022] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Immune-inflammatory response plays a key role in the pathogenesis of atherosclerosis. IgG N-glycosylation is reported to be associated with the 10-year atherosclerotic cardiovascular disease risk score and subclinical atherosclerosis. However, the relationship of IgG glycosylation with actual clinical cardiovascular disease (CVD) events and plaque phenotypes has rarely been investigated. Therefore, this study aims to understand whether IgG glycosylation traits are correlated with actual clinical CVD events and plaque phenotypes. METHODS AND ANALYSIS Designed to verify the efficacy of IgG glycosylation as a risk for CVD events and screen potential biomarkers of CVD to prevent atherosclerosis occurrence, this longitudinal prospective cohort study will be conducted at the First Affiliated Hospital of Shantou University Medical College, China. In total, 2720 participants routinely examined by carotid ultrasound will be divided into different groups according to plaque phenotype characteristics. Ultra-performance liquid chromatography will be performed to separate and detect IgG N-glycans in serum collected at baseline and at the end of the first, second and third years. The primary outcome is the actual clinical CVD composite events, including non-fatal myocardial infarction, death due to coronary heart disease, and fatal or non-fatal stroke. ETHICS AND DISSEMINATION The Clinical Ethics Committee of the First Affiliated Hospital of Shantou University Medical College approved this study (number: B-2021-127). Findings of this study will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER ChiCTR2100048740.
Collapse
Affiliation(s)
- Cuihong Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Centre for Precision Health, Edith Cowan University, Perth, Western Australia, Australia
| | - Gehendra Mahara
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hongxia Zhang
- Health Care Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xuerui Tan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
21
|
Zhang Z, Qian H, Tao Z, Xie Y, Zhi S, Sheng L, He W, Zhang L. Circulating circular RNAs as biomarkers for the diagnosis of essential hypertension with carotid plaque. Clin Exp Hypertens 2022; 44:601-609. [PMID: 35787223 DOI: 10.1080/10641963.2022.2093894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND At present, no early diagnostic markers for essential hypertension (EH)-induced subclinical target organs damage (such as carotid plaque) are available. This study aimed to identify the circular RNAs (circRNAs) in EH with carotid plaques, and assess their utility as biomarkers. METHODS First, circRNAs were identified through microarry analysis and database prediction. Second, a case-control study of EH patients with carotid plaque (n = 100) and healthy controls (n = 100) was performed to evaluate circRNAs expression in peripheral blood. Finally, receiver operating characteristic (ROC) curve was established to evaluate the diagnostic value. RESULTS Five circRNAs (hsa_circ_0105130, hsa_circ_0109569, hsa_circ_0072659, hsa_circ_0079586 and hsa_circ_0064684) were identified as the candidate circRNAs. We found that circRNAs were increased in case group compared with controls (P < .05). The results of ROC shown that these five circRNAs, especially hsa_circ_0109569 (AUC = 0.741), all had the moderate predictive value. CONCLUSIONS Our study revealed circulating circRNAs may act as promising noninvasive biomarkers for early detection and population screening of EH-induced subclinical target organ injury.
Collapse
Affiliation(s)
- Zebo Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, ZJ, China
| | - Haiyan Qian
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, ZJ, China
| | - Zhenbo Tao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, ZJ, China
| | - Yanqing Xie
- Institute of Geriatrics, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, ZJ, China
| | - Shuai Zhi
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, ZJ, China
| | - Liufang Sheng
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, ZJ, China
| | - Wenming He
- Institute of Geriatrics, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, ZJ, China
| | - Lina Zhang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, ZJ, China.,Institute of Geriatrics, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, ZJ, 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
|
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
|
24
|
Lu Y, Sun Y, Xu K, Saaoud F, Shao Y, Drummer C, Wu S, Hu W, Yu J, Kunapuli SP, Bethea JR, Vazquez-Padron RI, Sun J, Jiang X, Wang H, Yang X. Aorta in Pathologies May Function as an Immune Organ by Upregulating Secretomes for Immune and Vascular Cell Activation, Differentiation and Trans-Differentiation-Early Secretomes may Serve as Drivers for Trained Immunity. Front Immunol 2022; 13:858256. [PMID: 35320939 PMCID: PMC8934864 DOI: 10.3389/fimmu.2022.858256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023] Open
Abstract
To determine whether aorta becomes immune organ in pathologies, we performed transcriptomic analyses of six types of secretomic genes (SGs) in aorta and vascular cells and made the following findings: 1) 53.7% out of 21,306 human protein genes are classified into six secretomes, namely, canonical, caspase 1, caspase 4, exosome, Weibel-Palade body, and autophagy; 2) Atherosclerosis (AS), chronic kidney disease (CKD) and abdominal aortic aneurysm (AAA) modulate six secretomes in aortas; and Middle East Respiratory Syndrome Coronavirus (MERS-CoV, COVID-19 homologous) infected endothelial cells (ECs) and angiotensin-II (Ang-II) treated vascular smooth muscle cells (VSMCs) modulate six secretomes; 3) AS aortas upregulate T and B cell immune SGs; CKD aortas upregulate SGs for cardiac hypertrophy, and hepatic fibrosis; and AAA aorta upregulate SGs for neuromuscular signaling and protein catabolism; 4) Ang-II induced AAA, canonical, caspase 4, and exosome SGs have two expression peaks of high (day 7)-low (day 14)-high (day 28) patterns; 5) Elastase induced AAA aortas have more inflammatory/immune pathways than that of Ang-II induced AAA aortas; 6) Most disease-upregulated cytokines in aorta may be secreted via canonical and exosome secretomes; 7) Canonical and caspase 1 SGs play roles at early MERS-CoV infected ECs whereas caspase 4 and exosome SGs play roles in late/chronic phases; and the early upregulated canonical and caspase 1 SGs may function as drivers for trained immunity (innate immune memory); 8) Venous ECs from arteriovenous fistula (AVF) upregulate SGs in five secretomes; and 9) Increased some of 101 trained immunity genes and decreased trained tolerance regulator IRG1 participate in upregulations of SGs in atherosclerotic, Ang-II induced AAA and CKD aortas, and MERS-CoV infected ECs, but less in SGs upregulated in AVF ECs. IL-1 family cytokines, HIF1α, SET7 and mTOR, ROS regulators NRF2 and NOX2 partially regulate trained immunity genes; and NRF2 plays roles in downregulating SGs more than that of NOX2 in upregulating SGs. These results provide novel insights on the roles of aorta as immune organ in upregulating secretomes and driving immune and vascular cell differentiations in COVID-19, cardiovascular diseases, inflammations, transplantations, autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Yifan Lu
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Yu Sun
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Keman Xu
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Fatma Saaoud
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ying Shao
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Charles Drummer
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Sheng Wu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wenhui Hu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Jun Yu
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Satya P. Kunapuli
- Sol Sherry Thrombosis Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - John R. Bethea
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, United States
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Cardiovascular Research Center, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
- Center for Metabolic Disease Research, Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
25
|
Keeter WC, Ma S, Stahr N, Moriarty AK, Galkina EV. Atherosclerosis and multi-organ-associated pathologies. Semin Immunopathol 2022; 44:363-374. [PMID: 35238952 PMCID: PMC9069968 DOI: 10.1007/s00281-022-00914-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the vascular system that is characterized by the deposition of modified lipoproteins, accumulation of immune cells, and formation of fibrous tissue within the vessel wall. The disease occurs in vessels throughout the body and affects the functions of almost all organs including the lymphoid system, bone marrow, heart, brain, pancreas, adipose tissue, liver, kidneys, and gastrointestinal tract. Atherosclerosis and associated factors influence these tissues via the modulation of local vascular functions, induction of cholesterol-associated pathologies, and regulation of local immune responses. In this review, we discuss how atherosclerosis interferers with functions of different organs via several common pathways and how the disturbance of immunity in atherosclerosis can result in disease-provoking dysfunctions in multiple tissues. Our growing appreciation of the implication of atherosclerosis and associated microenvironmental conditions in the multi-organ pathology promises to influence our understanding of CVD-associated disease pathologies and to provide new therapeutic opportunities.
Collapse
Affiliation(s)
- W Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Shelby Ma
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Natalie Stahr
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Alina K Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA.
| |
Collapse
|
26
|
Hoebinger C, Rajcic D, Hendrikx T. Oxidized Lipids: Common Immunogenic Drivers of Non-Alcoholic Fatty Liver Disease and Atherosclerosis. Front Cardiovasc Med 2022; 8:824481. [PMID: 35083304 PMCID: PMC8784685 DOI: 10.3389/fcvm.2021.824481] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 12/17/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD), ranging from simple steatosis to inflammatory steatohepatitis (NASH) and cirrhosis, continues to rise, making it one of the major chronic liver diseases and indications for liver transplantation worldwide. The pathological processes underlying NAFLD not only affect the liver but are also likely to have systemic effects. In fact, growing evidence indicates that patients with NAFLD are at increased risk for developing atherosclerosis. Indeed, cardiovascular complications are the leading cause of mortality in NAFLD patients. Here, we aim to address common pathophysiological molecular pathways involved in chronic fatty liver disease and atherosclerosis. In particular, we focus on the role of oxidized lipids and the formation of oxidation-specific epitopes, which are important targets of host immunity. Acting as metabolic danger signals, they drive pro-inflammatory processes and thus contribute to disease progression. Finally, we summarize encouraging studies indicating that oxidized lipids are promising immunological targets to improve intervention strategies for NAFLD and potentially limit the risk of developing atherosclerosis.
Collapse
Affiliation(s)
- Constanze Hoebinger
- Department of Laboratory Medicine, Klinisches Institut für Labormedizin (KILM), Medical University Vienna, Vienna, Austria
| | - Dragana Rajcic
- Department of Laboratory Medicine, Klinisches Institut für Labormedizin (KILM), Medical University Vienna, Vienna, Austria
| | - Tim Hendrikx
- Department of Laboratory Medicine, Klinisches Institut für Labormedizin (KILM), Medical University Vienna, Vienna, Austria.,Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
27
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
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
|
Busch A, Bleichert S, Ibrahim N, Wortmann M, Eckstein HH, Brostjan C, Wagenhäuser MU, Goergen CJ, Maegdefessel L. Translating mouse models of abdominal aortic aneurysm to the translational needs of vascular surgery. JVS Vasc Sci 2021; 2:219-234. [PMID: 34778850 PMCID: PMC8577080 DOI: 10.1016/j.jvssci.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/04/2021] [Indexed: 01/03/2023] Open
Abstract
Introduction Abdominal aortic aneurysm (AAA) is a condition that has considerable socioeconomic impact and an eventual rupture is associated with high mortality and morbidity. Despite decades of research, surgical repair remains the treatment of choice and no medical therapy is currently available. Animal models and, in particular, murine models, of AAA are a vital tool for experimental in vivo research. However, each of the different models has individual limitations and provide only partial mimicry of human disease. This narrative review addresses the translational potential of the available mouse models, highlighting unanswered questions from a clinical perspective. It is based on a thorough presentation of the available literature and more than a decade of personal experience, with most of the available models in experimental and translational AAA research. Results From all the models published, only the four inducible models, namely the angiotensin II model (AngII), the porcine pancreatic elastase perfusion model (PPE), the external periadventitial elastase application (ePPE), and the CaCl2 model have been widely used by different independent research groups. Although the angiotensin II model provides features of dissection and aneurysm formation, the PPE model shows reliable features of human AAA, especially beyond day 7 after induction, but remains technically challenging. The translational value of ePPE as a model and the combination with β-aminopropionitrile to induce rupture and intraluminal thrombus formation is promising, but warrants further mechanistic insights. Finally, the external CaCl2 application is known to produce inflammatory vascular wall thickening. Unmet translational research questions include the origin of AAA development, monitoring aneurysm growth, gender issues, and novel surgical therapies as well as novel nonsurgical therapies. Conclusion New imaging techniques, experimental therapeutic alternatives, and endovascular treatment options provide a plethora of research topics to strengthen the individual features of currently available mouse models, creating the possibility of shedding new light on translational research questions.
Collapse
Affiliation(s)
- Albert Busch
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany.,Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Berlin, Germany
| | - Sonja Bleichert
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Nahla Ibrahim
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus Wortmann
- Department of Vascular and Endovascular Surgery, Universitaetsklinik Heidelberg, Heidelberg, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Christine Brostjan
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus U Wagenhäuser
- Department of Vascular and Endovascular Surgery, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Ind
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany.,Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Berlin, Germany
| |
Collapse
|
30
|
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
|
31
|
Reiche ME, Lutgens E. Lose the helpers… get 'remote' regulators! Cardiovasc Res 2021; 117:635-636. [PMID: 32637996 DOI: 10.1093/cvr/cvaa197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Myrthe E Reiche
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Pettenkoferstraße 8a, 80336 Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
32
|
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
|
33
|
Hutchinson MA, Park HS, Zanotti KJ, Alvarez-Gonzalez J, Zhang J, Zhang L, Telljohann R, Wang M, Lakatta EG, Gearhart PJ, Maul RW. Auto-Antibody Production During Experimental Atherosclerosis in ApoE-/- Mice. Front Immunol 2021; 12:695220. [PMID: 34305930 PMCID: PMC8299997 DOI: 10.3389/fimmu.2021.695220] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
Current models stipulate that B cells and antibodies function during atherosclerosis in two distinct ways based on antibody isotype, where IgM is protective and IgG is inflammatory. To examine this model, we generated ApoE-/- Aid-/- mice, which are unable to produce IgG antibodies due to the absence of activation-induced deaminase (AID) but maintain high plasma cholesterol due to the absence of apolipoprotein E (APOE). We saw a dramatic decrease in plaque formation in ApoE-/- Aid-/- mice compared to ApoE-/- mice. Rigorous analysis of serum antibodies revealed both ApoE-/- and ApoE-/- Aid-/- mice had substantially elevated titers of IgM antibodies compared to C57BL/6J controls, suggesting a more complex dynamic than previously described. Analysis of antigen specificity demonstrated that ApoE-/- Aid-/- mice had elevated titers of antibodies specific to malondialdehyde-oxidized low density lipoprotein (MDA-oxLDL), which has been shown to block macrophage recruitment into plaques. Conversely, ApoE-/- mice showed low levels of MDA-oxLDL specificity, but had antibodies specific to numerous self-proteins. We provide evidence for a hierarchical order of antibody specificity, where elevated levels of MDA-oxLDL specific IgM antibodies inhibit plaque formation. If the level of MDA-oxLDL specific IgM is insufficient, self-reactive IgM and IgG antibodies are generated against debris within the arterial plaque, resulting in increased inflammation and further plaque expansion.
Collapse
Affiliation(s)
- Mark A. Hutchinson
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Han-Sol Park
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Kimberly J. Zanotti
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Juan Alvarez-Gonzalez
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Jing Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Li Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Richard Telljohann
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Patricia J. Gearhart
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Robert W. Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| |
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: 45] [Impact Index Per Article: 11.3] [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
|
Cell-specific and divergent roles of the CD40L-CD40 axis in atherosclerotic vascular disease. Nat Commun 2021; 12:3754. [PMID: 34145241 PMCID: PMC8213756 DOI: 10.1038/s41467-021-23909-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/19/2021] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a major underlying cause of cardiovascular disease. Previous studies showed that inhibition of the co-stimulatory CD40 ligand (CD40L)-CD40 signaling axis profoundly attenuates atherosclerosis. As CD40L exerts multiple functions depending on the cell-cell interactions involved, we sought to investigate the function of the most relevant CD40L-expressing cell types in atherosclerosis: T cells and platelets. Atherosclerosis-prone mice with a CD40L-deficiency in CD4+ T cells display impaired Th1 polarization, as reflected by reduced interferon-γ production, and smaller atherosclerotic plaques containing fewer T-cells, smaller necrotic cores, an increased number of smooth muscle cells and thicker fibrous caps. Mice with a corresponding CD40-deficiency in CD11c+ dendritic cells phenocopy these findings, suggesting that the T cell-dendritic cell CD40L-CD40 axis is crucial in atherogenesis. Accordingly, sCD40L/sCD40 and interferon-γ concentrations in carotid plaques and plasma are positively correlated in patients with cerebrovascular disease. Platelet-specific deficiency of CD40L does not affect atherogenesis but ameliorates atherothrombosis. Our results establish divergent and cell-specific roles of CD40L-CD40 in atherosclerosis, which has implications for therapeutic strategies targeting this pathway.
Collapse
|
36
|
Meng Y, Zhang C, Liang L, Wei L, Wang H, Zhou F, Li R, Zou D, Huang X, Liu J. Identification of Potential Key Genes Involved in the Carotid Atherosclerosis. Clin Interv Aging 2021; 16:1071-1084. [PMID: 34140767 PMCID: PMC8203271 DOI: 10.2147/cia.s312941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Carotid atherosclerosis is a kind of systemic atherosclerosis in the carotid arteries. However, the efficiency of treatment is insufficient. Therefore, it is urgent to find therapeutic targets and deepen the understanding of carotid atherosclerosis. Materials and Methods In this study, we analyzed differentially expressed genes (DEGs) between atheroma plaque and macroscopically intact tissue (control samples). Furthermore, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes (KEGG) enrichment analysis based on the DEGs. Four methods were used to identify the hub genes in the protein–protein interaction networks of the DEGs. Furthermore, we also performed network module analysis to reveal carotid atherosclerosis-related gene modules and biological functions. Results The enrichment results showed that the biological functions were related to inflammation, immunity, chemokine and cell adhesion molecule, such as PIK-Akt signaling pathway, Rap1 signaling pathway, MAPK signaling pathway, NOD-like receptor signaling pathway and B cell receptor signaling pathway. In addition, we screened the hub genes. A total of 16 up-regulated genes (C3AR1, CCR1, CCR2, CD33, CD53, CXCL10, CXCL8, CXCR4, CYBB, FCER1G, FPR2, ITGAL, ITGAM, ITGAX, ITGB2, and LILRB2) were identified as hub genes. A total of 5 gene modules were obtained. We found that biological functions obtained for each cluster were mostly related to immunity, chemokines and cell adhesion molecules. Conclusion The present study identified key DEGs in atheroma plaque compared with control samples. The key genes involved in the development of carotid atherosclerosis may provide valuable therapeutic targets for carotid atherosclerosis.
Collapse
Affiliation(s)
- Youshi Meng
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Chunli Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Lucong Liang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Lei Wei
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Hao Wang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Fengkun Zhou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Rongjie Li
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China.,Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi, 530022, People's Republic of China
| | - Xiaohua Huang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, People's Republic of China
| | - Jie Liu
- Department of Cardiology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| |
Collapse
|
37
|
Abstract
Graphical abstract Establishment of an autoreactive B cell memory after myocardial infarction: a working hypothesis. The demise of cardiac cells leads to the release of cryptoantigens that induce a humoral immune response that leads to accumulation of immunoglobulins in plaques and eventually amplifies atherogenesis at remote sites.
Collapse
Affiliation(s)
- Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford
| | - Jennifer Cole
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford
| |
Collapse
|
38
|
Zhao Y, Chen W, Liu Y, Li H, Chi J, Chang Q, Shen L, Yan R, Li J, Yin X, Fu Y. Promoting plaque stability by gene silencing of monocyte chemotactic protein-3 or overexpression of tissue factor pathway inhibitor in ApoE-/- mice. J Drug Target 2021; 29:669-675. [PMID: 33472448 DOI: 10.1080/1061186x.2021.1878363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chemokines may promote the formation and instability of atherosclerotic plaque, which is the most common cause of acute coronary syndrome. The aim of this study was to clarify the function of monocyte chemotactic protein-3 (MCP-3) in the stability of atherosclerotic plaque, to determine the role of tissue factor pathway inhibitor (TFPI) on the development and stability of atherosclerotic plaques, and to further elucidate the anti-atherosclerotic mechanism of TFPI with the emphasis on chemokine MCP-3. We constructed an adenovirus-mediated shRNA against mouse MCP-3 (Ad-MCP-3-shRNA) and an adenovirus-containing TFPI (Ad-TFPI), and tranferred them in a model of vulnerable plaque in ApoE-/- mice respectively. Here, we reported that MCP-3-shRNA and TFPI could both reduce the plaque area and decrease the content of lipids and macrophages, on the contrary, the fibrous cap thickness and content of collagen and smooth muscle cells were increased. In addition, the expression of MCP-3 and CC chemokine receptor 2 (CCR2) was decreased by TFPI transfer. These data provide the first in vivo evidence that MCP-3 is a major contributor to the unstability of atherosclerotic plaque and TFPI may exert its anti-atherosclerotic effects and promote stabilisation of plaque at least partly through inhibiting MCP-3/CCR2 pathway, which may be a new therapeutic method for atherosclerosis.
Collapse
Affiliation(s)
- Yong Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Li
- Department of Cardiology, The Fifth Hospital in Harbin, Harbin, China
| | - Jinyu Chi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing Chang
- Department of Cardiology, Heilongjiang Provincial Hospital, Harbin, China
| | - Li Shen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Runan Yan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiashu Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhua Yin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
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
|
40
|
Lorenzo C, Delgado P, Busse CE, Sanz-Bravo A, Martos-Folgado I, Bonzon-Kulichenko E, Ferrarini A, Gonzalez-Valdes IB, Mur SM, Roldán-Montero R, Martinez-Lopez D, Martin-Ventura JL, Vázquez J, Wardemann H, Ramiro AR. ALDH4A1 is an atherosclerosis auto-antigen targeted by protective antibodies. Nature 2020; 589:287-292. [PMID: 33268892 DOI: 10.1038/s41586-020-2993-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in the world, with most CVD-related deaths resulting from myocardial infarction or stroke. The main underlying cause of thrombosis and cardiovascular events is atherosclerosis, an inflammatory disease that can remain asymptomatic for long periods. There is an urgent need for therapeutic and diagnostic options in this area. Atherosclerotic plaques contain autoantibodies1,2, and there is a connection between atherosclerosis and autoimmunity3. However, the immunogenic trigger and the effects of the autoantibody response during atherosclerosis are not well understood3-5. Here we performed high-throughput single-cell analysis of the atherosclerosis-associated antibody repertoire. Antibody gene sequencing of more than 1,700 B cells from atherogenic Ldlr-/- and control mice identified 56 antibodies expressed by in-vivo-expanded clones of B lymphocytes in the context of atherosclerosis. One-third of the expanded antibodies were reactive against atherosclerotic plaques, indicating that various antigens in the lesion can trigger antibody responses. Deep proteomics analysis identified ALDH4A1, a mitochondrial dehydrogenase involved in proline metabolism, as a target antigen of one of these autoantibodies, A12. ALDH4A1 distribution is altered during atherosclerosis, and circulating ALDH4A1 is increased in mice and humans with atherosclerosis, supporting the potential use of ALDH4A1 as a disease biomarker. Infusion of A12 antibodies into Ldlr-/- mice delayed plaque formation and reduced circulating free cholesterol and LDL, suggesting that anti-ALDH4A1 antibodies can protect against atherosclerosis progression and might have therapeutic potential in CVD.
Collapse
Affiliation(s)
- Cristina Lorenzo
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pilar Delgado
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Christian E Busse
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Alejandro Sanz-Bravo
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Elena Bonzon-Kulichenko
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Alessia Ferrarini
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ileana B Gonzalez-Valdes
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sonia M Mur
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Raquel Roldán-Montero
- Vascular Pathology Lab, IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Diego Martinez-Lopez
- Vascular Pathology Lab, IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Jose L Martin-Ventura
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Vascular Pathology Lab, IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Hedda Wardemann
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Almudena R Ramiro
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
41
|
Perrone MG, Miciaccia M, Vitale P, Ferorelli S, Araújo CDCB, de Almeida GS, Souza Domingos TF, da Silva LCRP, de Pádula M, Cabral LM, Sathler PC, Bonaccorso C, Fortuna CG, Scilimati A. An attempt to chemically state the cross-talk between monomers of COX homodimers by double/hybrid inhibitors mofezolac-spacer-mofezolac and mofezolac-spacer-arachidonic acid. Eur J Med Chem 2020; 209:112919. [PMID: 33129592 DOI: 10.1016/j.ejmech.2020.112919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases (CVDs) account for over 17 million death globally each year, including arterial thrombosis. Platelets are key components in the pathogenesis of this disease and modulating their activity is an effective strategy to treat such thrombotic events. Cyclooxygenase-1 (COX-1) isoenzyme is involved in platelet activation and is the main target of non-steroidal anti-inflammatory drugs (NSAIDs) and new selective inhibitor research. Inhibitors of general formula mofezolac-spacer-mofezolac (mof-spacer-mof) and mofezolac-spacer-arachidonic acid (mof-spacer-AA) were projected to investigate the possible cross-talk between the two monomers (Eallo and Ecat) forming the COX-1 homodimer. Mofezolac was chosen as either one or two moieties of these molecules being the known most potent and selective COX-1 inhibitor and administrated to humans as Disopain™, then arachidonic acid (AA) was used to develop molecules bearing, in the same compound, in addition to the inhibitor moiety (mofezolac) also the natural COX substrate. Depending on the nature of the spacer, COX-1 and COX-2 activity was differently inhibited by mof-spacer-mof set with a preferential COX-1 inhibition. The highest COX-1 selectivity was exhibited by the compound in which the spacer was the benzidine [N,N'-(biphenyl-4,4'-di-yl)bis (2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamide) (15): COX-1 IC50 = 0.08 μM, COX-2 IC50 > 50 μM, Selectivity Index (SI) > 625]. In the case of mof-spacer-AA set, the COX inhibitory potency and also the isoform preference changed. (5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-Bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}butyl)icosa-5,8,11,14-tetraenamide (19) and (5Z, 8Z, 11Z, 14Z)-N-(4'-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}-[1,1'-biphenyl]-4-yl)icosa-5,8,11,14-tetraenamide (21), in which the spacer is the 1,2-diaminobutane or benzidine, respectively, selectively inhibited the COX-2, whereas when the spacer is the 1,4-phenylendiamine [(5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}phenyl)icosa-5,8,11,14-tetraenamide) (20) the COX preference is COX-1 (COX-1 IC50 = 0.05 μM, COX-2 IC50 > 50 μM, with a COX-1 selectivity > 1000). Molecular modelling by using FLAP algorithm shows fundamental interactions of the novel compounds at the entry channel of COX and inside its catalytic cavity. The effect of these mof-spacer-mof and mof-spacer-AA in inhibiting in vitro free arachidonic acid-induced platelet aggregation was also determined. A positive profile of hemocompatibility in relation to their influence on the blood coagulation cascade and erythrocyte toxicity was observed. Cytotoxicity and genotoxicity safety were also found for these two novel sets of compounds.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Paola Vitale
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Cristina da Costa Bernardes Araújo
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Gabriella Silva de Almeida
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Thaisa Francielle Souza Domingos
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | | | - Marcelo de Pádula
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Plínio Cunha Sathler
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Carmela Bonaccorso
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Cosimo G Fortuna
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonio Scilimati
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
42
|
Karadimou G, Gisterå A, Gallina AL, Caravaca AS, Centa M, Salagianni M, Andreakos E, Hansson GK, Malin S, Olofsson PS, Paulsson-Berne G. Treatment with a Toll-like Receptor 7 ligand evokes protective immunity against atherosclerosis in hypercholesterolaemic mice. J Intern Med 2020; 288:321-334. [PMID: 32410352 DOI: 10.1111/joim.13085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The interplay between innate and adaptive immunity is central in life-threatening clinical complications of atherosclerosis such as myocardial infarction and stroke. The specific mechanisms involved and their protective versus detrimental effects in the disease process remain poorly understood. We have previously shown that higher levels of Toll-like receptor 7 (TLR7) expression in human atherosclerotic lesions are correlated with better patient outcome. OBJECTIVE In this study, we explored whether TLR7 activation can ameliorate disease in experimental atherosclerosis in mice. METHODS Apolipoprotein E deficient mice (Apoe-/- ) with established disease were injected for five weeks intraperitoneally with the TLR7 ligand R848. Local effects were evaluated by characterization of the lesion. Systemic effects of the treatment were investigated by immune composition analysis in the spleen and plasma measurements. RESULTS The in vivo treatment arrested lesion progression in the aorta. We also detected expansion of marginal zone B cells and Treg in the spleen together with increased plasma IgM antibodies against oxidized low-density lipoprotein (oxLDL) and reduced plasma cholesterol levels. These changes were accompanied by increased accumulation of IgM antibodies, decreased necrosis and fewer apoptotic cells in atherosclerotic lesions. CONCLUSIONS Our findings show that TLR7 stimulation could ameliorate atherosclerotic lesion burden and reduce plasma cholesterol in Apoe-/- mice. TLR7 stimulation was associated with an atheroprotective B-cell and Treg response, which may have systemic and local effects within lesions that could prevent arterial lipid accumulation and inflammation.
Collapse
Affiliation(s)
- G Karadimou
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - A Gisterå
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - A L Gallina
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - A S Caravaca
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - M Centa
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - M Salagianni
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - E Andreakos
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - G K Hansson
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - S Malin
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - P S Olofsson
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.,Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - G Paulsson-Berne
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
43
|
Roy P, Ali AJ, Kobiyama K, Ghosheh Y, Ley K. Opportunities for an atherosclerosis vaccine: From mice to humans. Vaccine 2020; 38:4495-4506. [PMID: 31964554 PMCID: PMC7939143 DOI: 10.1016/j.vaccine.2019.12.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/14/2023]
Abstract
Atherosclerosis, the major underlying cause of cardiovascular diseases (CVD), is the number one killer globally. The disease pathogenesis involves a complex interplay between metabolic and immune components. Although lipid-lowering drugs such as statins curb the risks associated with CVD, significant residual inflammatory risk remains. Substantial evidence from experimental models and clinical studies has established the role of inflammation and immune effector mechanisms in the pathogenesis of atherosclerosis. Several stages of the disease are affected by host-mediated antigen-specific adaptive immune responses that play either protective or proatherogenic roles. Therefore, strategies to boost an anti-atherogenic humoral and T regulatory cell response are emerging as preventative or therapeutic strategies to lowering inflammatory residual risks. Vaccination holds promise as an efficient, durable and relatively inexpensive approach to induce protective adaptive immunity in atherosclerotic patients. In this review, we discuss the status and opportunities for a human atherosclerosis vaccine. We describe (1) some of the immunomodulatory therapeutic interventions tested in atherosclerosis (2) the immune targets identified in pre-clinical and clinical investigations (3) immunization strategies evaluated in animal models (4) past and ongoing clinical trials to examine the safety and efficacy of human atherosclerosis vaccines and (5) strategies to improve and optimize vaccination in humans (antigen selection, formulation, dose and delivery).
Collapse
Affiliation(s)
- Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420, Athena Circle Drive, La Jolla, CA 92037, USA
| | - Amal J Ali
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420, Athena Circle Drive, La Jolla, CA 92037, USA
| | - Kouji Kobiyama
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420, Athena Circle Drive, La Jolla, CA 92037, USA; Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yanal Ghosheh
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420, Athena Circle Drive, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420, Athena Circle Drive, La Jolla, CA 92037, USA; Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, MC0412, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Ståhle M, Silvola JMU, Hellberg S, de Vries M, Quax PHA, Kroon J, Rinne P, de Jong A, Liljenbäck H, Savisto N, Wickman A, Stroes ESG, Ylä-Herttuala S, Saukko P, Abrahamsson T, Pettersson K, Knuuti J, Roivainen A, Saraste A. Therapeutic Antibody Against Phosphorylcholine Preserves Coronary Function and Attenuates Vascular 18F-FDG Uptake in Atherosclerotic Mice. JACC Basic Transl Sci 2020; 5:360-373. [PMID: 32368695 PMCID: PMC7188869 DOI: 10.1016/j.jacbts.2020.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/17/2022]
Abstract
Phosphorylcholine is a pro-inflammatory epitope in atherogenic oxidized phospholipids. This study investigated effects of a novel monoclonal IgG1 antibody against PC on vascular function and atherosclerotic inflammation. Treatment with phosphorylcholine antibody preserved coronary flow reserve and decreased uptake of 18F-FDG in atherosclerotic lesions in hypercholesterolemic mice. Noninvasive imaging techniques represent translational tools to assess the efficacy of phosphorylcholine-targeted therapy on coronary artery function and atherosclerosis.
This study showed that treatment with a therapeutic monoclonal immunoglobulin-G1 antibody against phosphorylcholine on oxidized phospholipids preserves coronary flow reserve and attenuates atherosclerotic inflammation as determined by the uptake of 18F-fluorodeoxyglucose in atherosclerotic mice. The noninvasive imaging techniques represent translational tools to assess the efficacy of phosphorylcholine-targeted therapy on coronary artery function and atherosclerosis in clinical studies.
Collapse
Key Words
- 18F-FDG, 18F-fluorodeoxyglucose
- 18F-fluorodeoxyglucose positron emission tomography
- ANOVA, analysis of variance
- ApoB, apolipoprotein-B
- CFR, coronary flow reserve
- HAEC, human aortic endothelial cell
- ICAM, intracellular adhesion molecule
- IL, interleukin
- Ig, immunoglobulin
- LDLR, low-density lipoprotein receptor
- Lp(a), lipoprotein(a)
- NO, nitric oxide
- OxLDL, oxidized low-density lipoprotein cholesterol
- OxPLs, oxidized phospholipids
- PC, phosphorylcholine
- PC-mAb, human PC antibody
- VCAM, vascular cell adhesion molecule
- atherosclerosis
- coronary flow reserve
- inflammation
- phosphorylcholine
Collapse
Affiliation(s)
- Mia Ståhle
- Turku PET Centre, University of Turku, Turku, Finland
| | | | | | - Margreet de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Petteri Rinne
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Alwin de Jong
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Nina Savisto
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Erik S G Stroes
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands.,Department of Vascular Medicine, Academic Medical Center, Amsterdam University Medical Center (UMC), Amsterdam, the Netherlands
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pekka Saukko
- Department of Pathology and Forensic Medicine, University of Turku, Turku, Finland
| | | | | | - Juhani Knuuti
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,Heart Center, Turku University Hospital, Turku, Finland.,Institute of Clinical Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
45
|
|
46
|
Zhao L, Gimple RC, Yang Z, Wei Y, Gustafsson JÅ, Zhou S. Immunoregulatory Functions of Nuclear Receptors: Mechanisms and Therapeutic Implications. Trends Endocrinol Metab 2020; 31:93-106. [PMID: 31706690 DOI: 10.1016/j.tem.2019.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/29/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
Members of the nuclear receptor superfamily serve as master regulators in signaling by either positively or negatively regulating gene expression. Accumulating evidence has suggested that nuclear receptors are actively involved in immune responses, with specific roles in different immune cell compartments that contribute to both normal function and to disease development. The druggable properties of nuclear receptors have made them ideal modulatory therapeutic targets. Here, we revisit nuclear receptor biology, summarize recent advances in our understanding of the immunological functions of nuclear receptors, describe cell-type-specific roles and specific nuclear receptors in disease pathogenesis, and explore their potential as novel therapeutic targets. These nuclear receptor-dependent alterations in the immune system are amenable to pharmacological manipulation and suggest novel therapeutic strategies.
Collapse
Affiliation(s)
- Linjie Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, CA, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Zhengnan Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yuquan Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Center for Medical Innovation, Department of Biosciences and Nutrition at Novum, Karolinska Institute, Stockholm, Sweden.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
47
|
Caravaca AS, Centa M, Gallina AL, Tarnawski L, Olofsson PS. Neural reflex control of vascular inflammation. Bioelectron Med 2020; 6:3. [PMID: 32232111 PMCID: PMC7065709 DOI: 10.1186/s42234-020-0038-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/14/2020] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic inflammatory disease that underlies myocardial infarction and stroke. Efficacious treatment for hyperlipidemia and hypertension has significantly reduced morbidity and mortality in cardiovascular disease. However, atherosclerosis still confers a considerable risk of adverse cardiovascular events. In the current mechanistic understanding of the pathogenesis of atherosclerosis, inflammation is pivotal both in disease development and progression. Recent clinical data provided support for this notion and treatment targeting inflammation is currently being explored. Interestingly, neural reflexes regulate cytokine production and inflammation. Hence, new technology utilizing implantable devices to deliver electrical impulses to activate neural circuits are currently being investigated in treatment of inflammation. Hopefully, it may become possible to target vascular inflammation in cardiovascular disease using bioelectronic medicine. In this review, we discuss neural control of inflammation and the potential implications of new therapeutic strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- A. S. Caravaca
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - M. Centa
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - A. L. Gallina
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - L. Tarnawski
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - P. S. Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| |
Collapse
|
48
|
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: 25] [Impact Index Per Article: 5.0] [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
|
49
|
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
|
50
|
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
|