1
|
DeConne TM, Buckley DJ, Trott DW, Martens CR. The role of T cells in vascular aging, hypertension, and atherosclerosis. Am J Physiol Heart Circ Physiol 2024; 327:H1345-H1360. [PMID: 39423035 DOI: 10.1152/ajpheart.00570.2024] [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: 08/19/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Vascular dysfunction has emerged as a significant risk factor for the development of cardio- and cerebrovascular diseases (CVDs), which are currently the leading cause of morbidity and mortality worldwide. T lymphocytes (T cells) have been shown to be important modulators of vascular function in primary aging and CVDs, likely by producing inflammatory cytokines and reactive oxygen species that influence vasoprotective molecules. This review summarizes the role of T cells on vascular function in aging, hypertension, and atherosclerosis in animals and humans, and discusses potential T-cell targeted therapeutics to prevent, delay, or reverse vascular dysfunction.
Collapse
Affiliation(s)
- Theodore M DeConne
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - David J Buckley
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, United States
| | - Daniel W Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, United States
| | - Christopher R Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
2
|
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
|
3
|
Ait-Oufella H, Libby P. Inflammation and Atherosclerosis: Prospects for Clinical Trials. Arterioscler Thromb Vasc Biol 2024; 44:1899-1905. [PMID: 39167675 PMCID: PMC11343092 DOI: 10.1161/atvbaha.124.320155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Sorbonne Université, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Schäfer S, Gogiraju R, Rösch M, Kerstan Y, Beck L, Garbisch J, Saliba AE, Gisterå A, Hermanns HM, Boon L, Kastenmüller W, Schäfer K, Cochain C, Zernecke A. CD8 + T Cells Drive Plaque Smooth Muscle Cell Dedifferentiation in Experimental Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1852-1872. [PMID: 38868941 DOI: 10.1161/atvbaha.123.320084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Atherosclerosis is driven by the infiltration of the arterial intima by diverse immune cells and smooth muscle cells (SMCs). CD8+ T cells promote lesion growth during atherosclerotic lesion development, but their role in advanced atherosclerosis is less clear. Here, we studied the role of CD8+ T cells and their effects on SMCs in established atherosclerosis. METHODS CD8+ T cells were depleted in (SMC reporter) low-density lipoprotein receptor-deficient (Ldlr-/-) mice with established atherosclerotic lesions. Atherosclerotic lesion formation was examined, and single-cell RNA sequencing of aortic SMCs and their progeny was performed. Additionally, coculture experiments with primary aortic SMCs and CD8+ T cells were conducted. RESULTS Although we could not detect differences in atherosclerotic lesion size, an increased plaque SMC content was noted in mice after CD8+ T-cell depletion. Single-cell RNA sequencing of aortic lineage-traced SMCs revealed contractile SMCs and a modulated SMC cluster, expressing macrophage- and osteoblast-related genes. CD8+ T-cell depletion was associated with an increased contractile but decreased macrophage and osteoblast-like gene signature in this modulated aortic SMC cluster. Conversely, exposure of isolated aortic SMCs to activated CD8+ T cells decreased the expression of genes indicative of a contractile SMC phenotype and induced a macrophage and osteoblast-like cell state. Notably, CD8+ T cells triggered calcium deposits in SMCs under osteogenic conditions. Mechanistically, we identified transcription factors highly expressed in modulated SMCs, including Runx1, to be induced by CD8+ T cells in cultured SMCs in an IFNγ (interferon-γ)-dependent manner. CONCLUSIONS We here uncovered CD8+ T cells to control the SMC phenotype in atherosclerosis. CD8+ T cells promote SMC dedifferentiation and drive SMCs to adopt features of macrophage-like and osteoblast-like, procalcifying cell phenotypes. Given the critical role of SMCs in atherosclerotic plaque stability, CD8+ T cells could thus be explored as therapeutic target cells during lesion progression.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/immunology
- Cell Dedifferentiation
- Plaque, Atherosclerotic
- Mice
- Disease Models, Animal
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Cells, Cultured
- Male
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Phenotype
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- Aorta/pathology
- Aorta/immunology
- Aorta/metabolism
- Coculture Techniques
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
Collapse
Affiliation(s)
- Sarah Schäfer
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I, University Medicine Mainz, Germany (R.G., K.S.)
| | - Melanie Rösch
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Yvonne Kerstan
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Lina Beck
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Janine Garbisch
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Institute of Molecular Infection Biology Faculty of Medicine, University of Würzburg, Germany (A.-E.S.)
| | - Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (A.G.)
| | - Heike M Hermanns
- Medical Clinic II, Division of Hepatology (H.M.H.), University Hospital of Würzburg, Germany
| | | | | | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medicine Mainz, Germany (R.G., K.S.)
| | - Clément Cochain
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine (S.S., M.R., Y.K., L. Beck, J.G., C.C., A.Z.), University Hospital of Würzburg, Germany
| |
Collapse
|
5
|
Lu J, Meng J, Wu G, Wei W, Xie H, Liu Y. Th1 cells reduce the osteoblast-like phenotype in valvular interstitial cells by inhibiting NLRP3 inflammasome activation in macrophages. Mol Med 2024; 30:110. [PMID: 39080527 PMCID: PMC11287975 DOI: 10.1186/s10020-024-00882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND AND AIMS Inflammation is initiates the propagation phase of aortic valve calcification. The activation of NLRP3 signaling in macrophages plays a crucial role in the progression of calcific aortic valve stenosis (CAVS). IFN-γ regulates NLRP3 activity in macrophages. This study aimed to explore the mechanism of IFN-γ regulation and its impact on CAVS progression and valve interstitial cell transdifferentiation. METHODS AND RESULTS The number of Th1 cells and the expression of IFN-γ and STAT1 in the aortic valve, spleen and peripheral blood increased significantly as CAVS progressed. To explore the mechanisms underlying the roles of Th1 cells and IFN-γ, we treated CAVS mice with IFN-γ-AAV9 or an anti-IFN-γ neutralizing antibody. While IFN-γ promoted aortic valve calcification and dysfunction, it significantly decreased NLRP3 signaling in splenic macrophages and Ly6C+ monocytes. In vitro coculture showed that Th1 cells inhibited NLPR3 activation in ox-LDL-treated macrophages through the IFN-γR1/IFN-γR2-STAT1 pathway. Compared with untreated medium, conditioned medium from Th1-treated bone marrow-derived macrophages reduced the osteogenic calcification of valvular interstitial cells. CONCLUSION Inhibition of the NLRP3 inflammasome by Th1 cells protects against valvular interstitial cell calcification as a negative feedback mechanism of adaptive immunity toward innate immunity. This study provides a precision medicine strategy for CAVS based on the targeting of anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Jing Lu
- The First Clinical Medical College, Guangxi Medical University, Guangxi Zhuang Autonomous Region, Shuangyong Road 22, Nanning, 530021, P.R. China
| | - Jiaming Meng
- Department of Cardiology, Liuzhou People's Hospital, Guangxi, Zhuang Autonomous Region, Wenchang Road 8, Liuzhou, 545000, P.R. China
| | - Gang Wu
- Department of Cardiology, Liuzhou People's Hospital, Guangxi, Zhuang Autonomous Region, Wenchang Road 8, Liuzhou, 545000, P.R. China
| | - Wulong Wei
- Department of Cardiology, Liuzhou People's Hospital, Guangxi, Zhuang Autonomous Region, Wenchang Road 8, Liuzhou, 545000, P.R. China
| | - Huabao Xie
- The First Clinical Medical College, Guangxi Medical University, Guangxi Zhuang Autonomous Region, Shuangyong Road 22, Nanning, 530021, P.R. China.
| | - Yanli Liu
- Department of Cardiology, Liuzhou People's Hospital, Guangxi, Zhuang Autonomous Region, Wenchang Road 8, Liuzhou, 545000, P.R. China.
| |
Collapse
|
6
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
7
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2024:S2090-1232(24)00253-4. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [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: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
8
|
Ngai D, Sukka SR, Tabas I. Crosstalk between efferocytic myeloid cells and T-cells and its relevance to atherosclerosis. Front Immunol 2024; 15:1403150. [PMID: 38873597 PMCID: PMC11169609 DOI: 10.3389/fimmu.2024.1403150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
The interplay between myeloid cells and T-lymphocytes is critical to the regulation of host defense and inflammation resolution. Dysregulation of this interaction can contribute to the development of chronic inflammatory diseases. Important among these diseases is atherosclerosis, which refers to focal lesions in the arterial intima driven by elevated apolipoprotein B-containing lipoproteins, notably low-density lipoprotein (LDL), and characterized by the formation of a plaque composed of inflammatory immune cells, a collection of dead cells and lipids called the necrotic core, and a fibrous cap. As the disease progresses, the necrotic core expands, and the fibrous cap becomes thin, which increases the risk of plaque rupture or erosion. Plaque rupture leads to a rapid thrombotic response that can give rise to heart attack, stroke, or sudden death. With marked lowering of circulating LDL, however, plaques become more stable and cardiac risk is lowered-a process known as atherosclerosis regression. A critical aspect of both atherosclerosis progression and regression is the crosstalk between innate (myeloid cells) and adaptive (T-lymphocytes) immune cells. Myeloid cells are specialized at clearing apoptotic cells by a process called efferocytosis, which is necessary for inflammation resolution. In advanced disease, efferocytosis is impaired, leading to secondary necrosis of apoptotic cells, inflammation, and, most importantly, defective tissue resolution. In regression, efferocytosis is reawakened aiding in inflammation resolution and plaque stabilization. Here, we will explore how efferocytosing myeloid cells could affect T-cell function and vice versa through antigen presentation, secreted factors, and cell-cell contacts and how this cellular crosstalk may contribute to the progression or regression of atherosclerosis.
Collapse
Affiliation(s)
- David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Santosh R. Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Department of Physiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
9
|
Xu L, Chen F, Fan W, Saito S, Cao D. The role of γδT lymphocytes in atherosclerosis. Front Immunol 2024; 15:1369202. [PMID: 38774876 PMCID: PMC11106432 DOI: 10.3389/fimmu.2024.1369202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/18/2024] [Indexed: 05/24/2024] Open
Abstract
Atherosclerosis poses a significant threat to human health, impacting overall well-being and imposing substantial financial burdens. Current treatment strategies mainly focus on managing low-density lipids (LDL) and optimizing liver functions. However, it's crucial to recognize that Atherosclerosis involves more than just lipid accumulation; it entails a complex interplay of immune responses. Research highlights the pivotal role of lipid-laden macrophages in the formation of atherosclerotic plaques. These macrophages attract lymphocytes like CD4 and CD8 to the inflamed site, potentially intensifying the inflammatory response. γδ T lymphocytes, with their diverse functions in innate and adaptive immune responses, pathogen defense, antigen presentation, and inflammation regulation, have been implicated in the early stages of Atherosclerosis. However, our understanding of the roles of γδ T cells in Atherosclerosis remains limited. This mini-review aims to shed light on the characteristics and functions of γδ T cells in Atherosclerosis. By gaining insights into the roles of γδ T cells, we may uncover a promising strategy to mitigate plaque buildup and dampen the inflammatory response, thereby opening new avenues for effectively managing this condition.
Collapse
Affiliation(s)
- LiMin Xu
- Department of Neurosurgery, Shenzhen Entry-Exit Frontier Inspection Hospital, Shenzhen, China
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - DuoYao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
10
|
Vos WG, van Os BW, den Toom M, Beckers L, van Roomen CP, van Tiel CM, Mohapatra BC, Band H, Nitz K, Weber C, Atzler D, de Winther MP, Bosmans LA, Lutgens E, Seijkens TT. T cell specific deletion of Casitas B lineage lymphoma-b reduces atherosclerosis, but increases plaque T cell infiltration and systemic T cell activation. Front Immunol 2024; 15:1297893. [PMID: 38504977 PMCID: PMC10949527 DOI: 10.3389/fimmu.2024.1297893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Atherosclerosis is a lipid-driven inflammatory disease of the arterial wall, and the underlying cause of the majority of cardiovascular diseases. Recent advances in high-parametric immunophenotyping of immune cells indicate that T cells constitute the major leukocyte population in the atherosclerotic plaque. The E3 ubiquitin ligase Casitas B-lymphoma proto-oncogene-B (CBL-B) is a critical intracellular regulator that sets the threshold for T cell activation, making CBL-B a potential therapeutic target to modulate inflammation in atherosclerosis. We previously demonstrated that complete knock-out of CBL-B aggravated atherosclerosis in Apoe-/- mice, which was attributed to increased macrophage recruitment and increased CD8+ T cell activation in the plaque. Methods To further study the T cell specific role of CBL-B in atherosclerosis, Apoe-/- CD4cre Cblb fl/fl (Cbl-bcKO) mice and Apoe-/-CD4WTCblbfl/fl littermates (Cbl-bfl/fl) were fed a high cholesterol diet for ten weeks. Results Cbl-bcKO mice had smaller atherosclerotic lesions in the aortic arch and root compared to Cbl-bfl/fl, and a substantial increase in CD3+ T cells in the plaque. Collagen content in the plaque was decreased, while other plaque characteristics including plaque necrotic core, macrophage content, and smooth muscle cell content, remained unchanged. Mice lacking T cell CBL-B had a 1.4-fold increase in CD8+ T cells and a 1.8-fold increase in regulatory T cells in the spleen. Splenic CD4+ and CD8+ T cells had increased expression of C-X-C Motif Chemokine Receptor 3 (CXCR3) and interferon-γ (IFN-γ), indicating a T helper 1 (Th1)-like/effector CD8+ T cell-like phenotype. Conclusion In conclusion, Cbl-bcKO mice have reduced atherosclerosis but show increased T cell accumulation in the plaque accompanied by systemic T cell activation.
Collapse
Affiliation(s)
- Winnie G. Vos
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Bram W. van Os
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Cindy P.A.A. van Roomen
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
| | - Bhopal C. Mohapatra
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid Band
- Eppley Institute for Research in Cancer and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katrin Nitz
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, United States
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute of Parmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Laura A. Bosmans
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Inflammatory Diseases, Amsterdam, Netherlands
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, United States
| | - Tom T.P. Seijkens
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC) Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
11
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
12
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
13
|
Song J, Zhang Y, Frieler RA, Andren A, Wood S, Tyrrell DJ, Sajjakulnukit P, Deng JC, Lyssiotis CA, Mortensen RM, Salmon M, Goldstein DR. Itaconate suppresses atherosclerosis by activating a Nrf2-dependent antiinflammatory response in macrophages in mice. J Clin Invest 2023; 134:e173034. [PMID: 38085578 PMCID: PMC10849764 DOI: 10.1172/jci173034] [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: 06/22/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Itaconate has emerged as a critical immunoregulatory metabolite. Here, we examined the therapeutic potential of itaconate in atherosclerosis. We found that both itaconate and the enzyme that synthesizes it, aconitate decarboxylase 1 (Acod1, also known as immune-responsive gene 1 [IRG1]), are upregulated during atherogenesis in mice. Deletion of Acod1 in myeloid cells exacerbated inflammation and atherosclerosis in vivo and resulted in an elevated frequency of a specific subset of M1-polarized proinflammatory macrophages in the atherosclerotic aorta. Importantly, Acod1 levels were inversely correlated with clinical occlusion in atherosclerotic human aorta specimens. Treating mice with the itaconate derivative 4-octyl itaconate attenuated inflammation and atherosclerosis induced by high cholesterol. Mechanistically, we found that the antioxidant transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), was required for itaconate to suppress macrophage activation induced by oxidized lipids in vitro and to decrease atherosclerotic lesion areas in vivo. Overall, our work shows that itaconate suppresses atherogenesis by inducing Nrf2-dependent inhibition of proinflammatory responses in macrophages. Activation of the itaconate pathway may represent an important approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Ryan A. Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anthony Andren
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sherri Wood
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel J. Tyrrell
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Peter Sajjakulnukit
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center
| | - Jane C. Deng
- Graduate Program in Immunology, and
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard M. Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes
| | | | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Graduate Program in Immunology, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Tyrrell DJ, Wragg KM, Chen J, Wang H, Song J, Blin MG, Bolding C, Vardaman D, Giles K, Tidwell H, Ali MA, Janappareddi A, Wood SC, Goldstein DR. Clonally expanded memory CD8 + T cells accumulate in atherosclerotic plaques and are pro-atherogenic in aged mice. NATURE AGING 2023; 3:1576-1590. [PMID: 37996758 DOI: 10.1038/s43587-023-00515-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/27/2023] [Indexed: 11/25/2023]
Abstract
Aging is a strong risk factor for atherosclerosis and induces accumulation of memory CD8+ T cells in mice and humans. Biological changes that occur with aging lead to enhanced atherosclerosis, yet the role of aging on CD8+ T cells during atherogenesis is unclear. In this study, using femle mice, we found that depletion of CD8+ T cells attenuated atherogenesis in aged, but not young, animals. Furthermore, adoptive transfer of splenic CD8+ T cells from aged wild-type, but not young wild-type, donor mice significantly enhanced atherosclerosis in recipient mice lacking CD8+ T cells. We also characterized T cells in healthy and atherosclerotic young and aged mice by single-cell RNA sequencing. We found specific subsets of age-associated CD8+ T cells, including a Granzyme K+ effector memory subset, that accumulated and was clonally expanded within atherosclerotic plaques. These had transcriptomic signatures of T cell activation, migration, cytotoxicity and exhaustion. Overall, our study identified memory CD8+ T cells as therapeutic targets for atherosclerosis in aging.
Collapse
Affiliation(s)
- Daniel J Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Kathleen M Wragg
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jianrui Song
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Muriel G Blin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chase Bolding
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donald Vardaman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kara Giles
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harrison Tidwell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Sherri C Wood
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Daniel R Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Ma X, Zhuo Y, Huang Y, He P, Huang Z, Jiang L, Tong L, Yao X, Wen X, Zhong X, Yang S, Li C, Liu Y, Zhang Z. Reduced Diversities and Clonally Expanded Sequences of T-Cell Receptors in Patients With Essential Hypertension and Subclinical Carotid Atherosclerosis. Hypertension 2023; 80:2318-2329. [PMID: 37551594 DOI: 10.1161/hypertensionaha.123.21112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND It has long been hypothesized that the abnormal immune responses contribute to the essential hypertension (EH) and its subclinical target organ damage (STOD). However, the mechanism is unclear. This study aimed at exploring the potential association with abnormal T-cell responses and EH, STOD, and early atherosclerosis in patients with EH. METHODS This cross-sectional study included 146 patients with EH and 73 age-matched normotensive individuals. The expressed peripheral TCR (T-cell receptor) β repertoire was analyzed by high through-put sequencing. RESULTS The TCRβ repertoires of the patients with EH were significantly different, with significantly elevated certain TCR beta variable (TRBV) and joint (TRBJ) gene usages, significantly reduced TCRβ diversity indexes (diversity 50s) and numbers of total TCRβ clonal types, significantly elevated percentages of the biggest TCRβ clones and numbers of clones accounting >0.1% sequences, compared with those in the normotensive controls. Decreased diversity 50s and increased biggest TCRβ clone percentages were independently correlated with carotid intima-media thickness and subclinical carotid atherosclerosis (SCA) in the patients with EH. Moreover, the diversity 50s were further significantly reduced and the biggest TCRβ clone percentages were significantly increased in the patients with EH with SCA (n=89) comparing to the patients with EH/patients without SCA (n=57), and in patients with EH/SCA with carotid plaque (n=22) comparing to patients with EH/SCA/patients without carotid plaque (n=67). Importantly, specific TCRβ clones were identified in different subgroups of the patients with EH. CONCLUSIONS These results reveal that abnormal T-cell responses may play important roles in the progression of EH and its SCA, especially the formation of carotid plaque. REGISTRATION URL: https://www.chictr.org.cn; Unique identifier: ChiCTR2100054414.
Collapse
Affiliation(s)
- Xiaoxiang Ma
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Yue Zhuo
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Yu Huang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Pengming He
- Chengdu ExAb Biotechnology LTD, Chengdu, Sichuan, China (P.H., X.W., X.Z., S.Y., C.L.)
| | - Zhaoyi Huang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Li Jiang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Luyao Tong
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Xiaoqin Yao
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Xueping Wen
- Chengdu ExAb Biotechnology LTD, Chengdu, Sichuan, China (P.H., X.W., X.Z., S.Y., C.L.)
| | - Xuemei Zhong
- Chengdu ExAb Biotechnology LTD, Chengdu, Sichuan, China (P.H., X.W., X.Z., S.Y., C.L.)
| | - Shihan Yang
- Chengdu ExAb Biotechnology LTD, Chengdu, Sichuan, China (P.H., X.W., X.Z., S.Y., C.L.)
| | - Changqiong Li
- Chengdu ExAb Biotechnology LTD, Chengdu, Sichuan, China (P.H., X.W., X.Z., S.Y., C.L.)
| | - Yuping Liu
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| | - Zhixin Zhang
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan (X.M., Y.Z., Y.H., Z.H., L.J., L.T., X.Y., Y.L., Z.Z.)
| |
Collapse
|
16
|
Liu F, Wang Y, Yu J. Role of inflammation and immune response in atherosclerosis: Mechanisms, modulations, and therapeutic targets. Hum Immunol 2023; 84:439-449. [PMID: 37353446 DOI: 10.1016/j.humimm.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/19/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular diseases (CVDs) have emerged as the leading cause of mortality globally, with atherosclerosis being a prominent focus of investigation among medical researchers worldwide. Atherosclerosis is characterized as a disease of the large and medium-sized arteries that is multifocal, accumulative, and immunoinflammatory in nature, resulting from the deposition of lipids. Accumulating evidence suggests that inflammatory responses and immunoregulation play a vital role in the occurrence and development of atherosclerosis. While existing treatments for atherosclerosis can assist in symptom management and slowing disease progression, a complete cure remains elusive. Consequently, there is significant interest in research and development of potential new drugs for this condition. Therefore, this review aims to consolidate the current understanding of the pathogenesis of atherosclerosis with an emphasis on inflammation, immune response and infection. Besides, it examines the effects and mechanisms of immunological modulations in atherosclerosis, and the potential therapeutic targets and drugs for intervening in the inflammatory responses and immunoregulation associated with atherosclerosis. Additionally, novel drug options for treating atherosclerosis are explored within the context of this review.
Collapse
Affiliation(s)
- Fang Liu
- Department of Vascular Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| | - Yijun Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Jiayin Yu
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
17
|
Bazioti V, Halmos B, Westerterp M. T-cell Cholesterol Accumulation, Aging, and Atherosclerosis. Curr Atheroscler Rep 2023; 25:527-534. [PMID: 37395922 PMCID: PMC10471657 DOI: 10.1007/s11883-023-01125-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW The majority of leukocytes in advanced human atherosclerotic plaques are T-cells. T-cell subsets exert pro- or anti-atherogenic effects largely via the cytokines they secrete. Tregulatory cells (Tregs) are anti-inflammatory, but may lose these properties during atherosclerosis, proposed to be downstream of cholesterol accumulation. Aged T-cells also accumulate cholesterol. The effects of T-cell cholesterol accumulation on T-cell fate and atherosclerosis are not uniform. RECENT FINDINGS T-cell cholesterol accumulation enhances differentiation into pro-atherogenic cytotoxic T-cells and boosts their killing capacity, depending on the localization and extent of cholesterol accumulation. Excessive cholesterol accumulation induces T-cell exhaustion or T-cell apoptosis, the latter decreasing atherosclerosis but impairing T-cell functionality in terms of killing capacity and proliferation. This may explain the compromised T-cell functionality in aged T-cells and T-cells from CVD patients. The extent of T-cell cholesterol accumulation and its cellular localization determine T-cell fate and downstream effects on atherosclerosis and T-cell functionality.
Collapse
Affiliation(s)
- Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336, Munich, Germany
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands.
| |
Collapse
|
18
|
Jing J, Guo J, Dai R, Zhu C, Zhang Z. Targeting gut microbiota and immune crosstalk: potential mechanisms of natural products in the treatment of atherosclerosis. Front Pharmacol 2023; 14:1252907. [PMID: 37719851 PMCID: PMC10504665 DOI: 10.3389/fphar.2023.1252907] [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: 07/04/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory reaction that primarily affects large and medium-sized arteries. It is a major cause of cardiovascular disease and peripheral arterial occlusive disease. The pathogenesis of AS involves specific structural and functional alterations in various populations of vascular cells at different stages of the disease. The immune response is involved throughout the entire developmental stage of AS, and targeting immune cells presents a promising avenue for its treatment. Over the past 2 decades, studies have shown that gut microbiota (GM) and its metabolites, such as trimethylamine-N-oxide, have a significant impact on the progression of AS. Interestingly, it has also been reported that there are complex mechanisms of action between GM and their metabolites, immune responses, and natural products that can have an impact on AS. GM and its metabolites regulate the functional expression of immune cells and have potential impacts on AS. Natural products have a wide range of health properties, and researchers are increasingly focusing on their role in AS. Now, there is compelling evidence that natural products provide an alternative approach to improving immune function in the AS microenvironment by modulating the GM. Natural product metabolites such as resveratrol, berberine, curcumin, and quercetin may improve the intestinal microenvironment by modulating the relative abundance of GM, which in turn influences the accumulation of GM metabolites. Natural products can delay the progression of AS by regulating the metabolism of GM, inhibiting the migration of monocytes and macrophages, promoting the polarization of the M2 phenotype of macrophages, down-regulating the level of inflammatory factors, regulating the balance of Treg/Th17, and inhibiting the formation of foam cells. Based on the above, we describe recent advances in the use of natural products that target GM and immune cells crosstalk to treat AS, which may bring some insights to guide the treatment of AS.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Dai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chaojun Zhu
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
19
|
Hinkley H, Counts DA, VonCanon E, Lacy M. T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease. Cells 2023; 12:2152. [PMID: 37681883 PMCID: PMC10486666 DOI: 10.3390/cells12172152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we discuss the recognition of atherosclerosis-related antigens that may trigger T cell activation together with additional signaling from co-stimulatory molecules and lesional cytokines. Although few studies have indicated candidates for the antigen specificity of T cells in atherosclerosis, further research is needed. Furthermore, we describe the pro-atherogenic and atheroprotective roles of diverse subsets of T cells such as CD4+ helper, CD8+ cytotoxic, invariant natural killer, and γδ T cells. To classify and quantify T cell subsets in atherosclerosis, we summarize current methods to analyze cellular heterogeneity including single cell RNA sequencing and T cell receptor (TCR) sequencing. Further insights into T cell biology will help shed light on the immunopathology of atherosclerosis, inform potential therapeutic interventions, and pave the way for precision medicine approaches in combating cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Michael Lacy
- Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
20
|
Jing J, Zhu C, Gong R, Qi X, Zhang Y, Zhang Z. Research progress on the active ingredients of traditional Chinese medicine in the intervention of atherosclerosis: A promising natural immunotherapeutic adjuvant. Biomed Pharmacother 2023; 159:114201. [PMID: 36610225 DOI: 10.1016/j.biopha.2022.114201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease caused by disorders of lipid metabolism. Abnormal deposition of low-density lipoproteins in the arterial wall stimulates the activation of immune cells, including the adhesion and infiltration of monocytes, the proliferation and differentiation of macrophages and lymphocytes, and the activation of their functions. The complex interplay between immune cells coordinates the balance between pro- and anti-inflammation and plays a key role in the progression of AS. Therefore, targeting immune cell activity may lead to the development of more selective drugs with fewer side effects to treat AS without compromising host defense mechanisms. At present, an increasing number of studies have found that the active ingredients of traditional Chinese medicine (TCM) can regulate the function of immune cells in multiple ways to against AS, showing great potential for the treatment of AS and promising clinical applications. In this paper, we review the mechanisms of immune cell action in AS lesions and the potential targets and/or pathways for immune cell regulation by the active ingredients of TCM to promote the understanding of the immune system interactions of AS and provide a relevant basis for the use of active ingredients of TCM as natural adjuvants for AS immunotherapy.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Chaojun Zhu
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Rui Gong
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Xue Qi
- Department of General Surgery, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China.
| | - Yue Zhang
- Peripheral Vascular Disease Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhaohui Zhang
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
21
|
Zhu X, Li Q, George V, Spanoudis C, Gilkes C, Shrestha N, Liu B, Kong L, You L, Echeverri C, Li L, Wang Z, Chaturvedi P, Muniz GJ, Egan JO, Rhode PR, Wong HC. A novel interleukin-2-based fusion molecule, HCW9302, differentially promotes regulatory T cell expansion to treat atherosclerosis in mice. Front Immunol 2023; 14:1114802. [PMID: 36761778 PMCID: PMC9907325 DOI: 10.3389/fimmu.2023.1114802] [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: 12/06/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused by deposition of oxidative low-density lipoprotein (LDL) in the arterial intima which triggers the innate immune response through myeloid cells such as macrophages. Regulatory T cells (Tregs) play an important role in controlling the progression or regression of atherosclerosis by resolving macrophage-mediated inflammatory functions. Interleukin-2 (IL-2) signaling is essential for homeostasis of Tregs. Since recombinant IL-2 has an unfavorable pharmacokinetic profile limiting its therapeutic use, we constructed a fusion protein, designated HCW9302, containing two IL-2 domains linked by an extracellular tissue factor domain. We found that HCW9302 exhibited a longer serum half-life with an approximately 1000-fold higher affinity for the IL-2Rα than IL-2. HCW9302 could be administered to mice at a dosing range that expanded and activated Tregs but not CD4+ effector T cells. In an ApoE-/- mouse model, HCW9302 treatment curtailed the progression of atherosclerosis through Treg activation and expansion, M2 macrophage polarization and myeloid-derived suppressor cell induction. HCW9302 treatment also lessened inflammatory responses in the aorta. Thus, HCW9302 is a potential therapeutic agent to expand and activate Tregs for treatment of inflammatory and autoimmune diseases.
Collapse
|
22
|
A Single-Cell Atlas of the Atherosclerotic Plaque in the Femoral Artery and the Heterogeneity in Macrophage Subtypes between Carotid and Femoral Atherosclerosis. J Cardiovasc Dev Dis 2022; 9:jcdd9120465. [PMID: 36547462 PMCID: PMC9788114 DOI: 10.3390/jcdd9120465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis of femoral arteries can cause the insufficient blood supply to the lower limbs and lead to gangrenous ulcers and other symptoms. Atherosclerosis and inflammatory factors are significantly different from other plaques. Therefore, it is crucial to observe the cellular composition of the femoral atherosclerotic plaque and identify plaque heterogeneity in other arteries. To this end, we performed single-cell sequencing of a human femoral artery plaque. We identified 14 cell types, including endothelial cells, smooth muscle cells, monocytes, three macrophages with four different subtypes of foam cells, three T cells, natural killer cells, and B cells. We then downloaded single-cell sequencing data of carotid atherosclerosis from GEO, which were compared with the one femoral sample. We identified similar cell types, but the femoral artery had significantly more nonspecific immune cells and fewer specific immune cells than the carotid artery. We further compared the differences in the proportion of inflammatory macrophages, and resident macrophages, and the proportion of inflammatory macrophages was greater within the carotid artery. Through comparing one femoral sequencing sample with carotid samples from public datasets, our study reveals the single-cell map of the femoral artery and the heterogeneity of carotid and femoral arteries at the cellular level, laying the foundation for mechanistic and pharmacological studies of the femoral artery.
Collapse
|
23
|
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: 6] [Impact Index Per Article: 2.0] [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
|
24
|
The Role of Hydrogen Sulfide in Plaque Stability. Antioxidants (Basel) 2022; 11:antiox11122356. [PMID: 36552564 PMCID: PMC9774534 DOI: 10.3390/antiox11122356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is the greatest contributor to cardiovascular events and is involved in the majority of deaths worldwide. Plaque rapture or erosion precipitates life-threatening thrombi, resulting in the obstruction blood flow to the heart (acute coronary syndrome), brain (ischemic stroke) or low extremities (peripheral vascular diseases). Among these events, major causation dues to the plaque rupture. Although the initiation, procession, and precise time of controlling plaque rupture are unclear, foam cell formation and apoptosis, cell death, extracellular matrix components, protease expression and activity, local inflammation, intraplaque hemorrhage, and calcification contribute to the plaque instability. These alterations tightly associate with the function regulation of intraplaque various cell populations. Hydrogen sulfide (H2S) is gasotransmitter derived from methionine metabolism and exerts a protective role in the genesis of atherosclerosis. Recent progress also showed H2S mediated the plaque stability. In this review, we discuss the progress of endogenous H2S modulation on functions of vascular smooth muscle cells, monocytes/macrophages, and T cells, and the molecular mechanism in plaque stability.
Collapse
|
25
|
Zhou Y, Wang S, Liang X, Heger Z, Xu M, Lu Q, Yu M, Adam V, Li N. Turning Hot into Cold: Immune Microenvironment Reshaping for Atherosclerosis Attenuation Based on pH-Responsive shSiglec-1 Delivery System. ACS NANO 2022; 16:10517-10533. [PMID: 35762565 DOI: 10.1021/acsnano.2c01778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Current atherosclerosis treatment is based on a combination of cholesterol-lowering medication and low-fat diets; however, the clinical effect is unsatisfactory. It has been shown that the level of immune cell infiltration and pro-inflammatory factors in the atherosclerotic immune microenvironment (AIM) play important roles in the development and progression of atherosclerosis. Therefore, we hypothesized that reshaping "hot AIM" into "cold AIM" could attenuate atherosclerosis. For this purpose, we designed a pH-responsive and charge-reversible nanosystem, referred to as Au-PEI/shSiglec-1/PEI-acetylsalicylic acid (ASPA NPs) to effectively deliver shSiglec-1, which blocked the interactions between macrophages with CD8+ T/NKT cells, thus inhibiting immune cell infiltration. Further, we demonstrated that acetylsalicylic acid (ASA), detached from the pH-responsive PEI-ASA polymer, and inhibited lipid accumulation in macrophage, thereby decreasing the lipid antigen presentation. Additionally, reduced macrophage-produced inflammatory factors by ASA and low CD8+ T/NKT cell infiltration levels synergistically inhibit Th17 cell differentiation, thus further dramatically attenuating inflammation in AIM by decreasing the IL-17A production. Eventually, ASPA NPs efficiently reshaped AIM by inhibiting immune cell infiltration, lipid antigen presentation, and pro-inflammation, which provided a feasible therapeutic strategy for atherosclerosis immunotherapy.
Collapse
Affiliation(s)
- Yue Zhou
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Siyu Wang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Qiang Lu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Meng Yu
- School of Pharmaceutical Science Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
26
|
Bazioti V, La Rose AM, Maassen S, Bianchi F, de Boer R, Halmos B, Dabral D, Guilbaud E, Flohr-Svendsen A, Groenen AG, Marmolejo-Garza A, Koster MH, Kloosterhuis NJ, Havinga R, Pranger AT, Langelaar-Makkinje M, de Bruin A, van de Sluis B, Kohan AB, Yvan-Charvet L, van den Bogaart G, Westerterp M. T cell cholesterol efflux suppresses apoptosis and senescence and increases atherosclerosis in middle aged mice. Nat Commun 2022; 13:3799. [PMID: 35778407 PMCID: PMC9249754 DOI: 10.1038/s41467-022-31135-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease driven by hypercholesterolemia. During aging, T cells accumulate cholesterol, potentially affecting inflammation. However, the effect of cholesterol efflux pathways mediated by ATP-binding cassette A1 and G1 (ABCA1/ABCG1) on T cell-dependent age-related inflammation and atherosclerosis remains poorly understood. In this study, we generate mice with T cell-specific Abca1/Abcg1-deficiency on the low-density-lipoprotein-receptor deficient (Ldlr-/-) background. T cell Abca1/Abcg1-deficiency decreases blood, lymph node, and splenic T cells, and increases T cell activation and apoptosis. T cell Abca1/Abcg1-deficiency induces a premature T cell aging phenotype in middle-aged (12-13 months) Ldlr-/- mice, reflected by upregulation of senescence markers. Despite T cell senescence and enhanced T cell activation, T cell Abca1/Abcg1-deficiency decreases atherosclerosis and aortic inflammation in middle-aged Ldlr-/- mice, accompanied by decreased T cells in atherosclerotic plaques. We attribute these effects to T cell apoptosis downstream of T cell activation, compromising T cell functionality. Collectively, we show that T cell cholesterol efflux pathways suppress T cell apoptosis and senescence, and induce atherosclerosis in middle-aged Ldlr-/- mice.
Collapse
Affiliation(s)
- Venetia Bazioti
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5252.00000 0004 1936 973XInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Anouk M. La Rose
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Sjors Maassen
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Frans Bianchi
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Rinse de Boer
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Benedek Halmos
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Deepti Dabral
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Emma Guilbaud
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Arthur Flohr-Svendsen
- grid.4494.d0000 0000 9558 4598European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Anouk G. Groenen
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alejandro Marmolejo-Garza
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Mirjam H. Koster
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Rick Havinga
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alle T. Pranger
- grid.4494.d0000 0000 9558 4598Laboratory of Medicine, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alain de Bruin
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5477.10000000120346234Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Bart van de Sluis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alison B. Kohan
- grid.21925.3d0000 0004 1936 9000Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Laurent Yvan-Charvet
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Geert van den Bogaart
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Marit Westerterp
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| |
Collapse
|
27
|
Lu X, Crowley SD. The Immune System in Hypertension: a Lost Shaker of Salt 2021 Lewis K. Dahl Memorial Lecture. Hypertension 2022; 79:1339-1347. [PMID: 35545942 DOI: 10.1161/hypertensionaha.122.18554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The seminal observations of Dr Lewis Dahl regarding renal mechanisms of hypertension remain highly relevant in light of more recent experiments showing that immune system dysfunction contributes to hypertension pathogenesis. Dr Dahl established that inappropriate salt retention in the kidney plays a central role via Ohm's Law in permitting blood pressure elevation. Nevertheless, inflammatory cytokines whose expression is induced in the early stages of hypertension can alter renal blood flow and sodium transporter expression and activity to foster renal sodium retention. By elaborating these cytokines and reactive oxygen species, myeloid cells and T lymphocytes can connect systemic inflammatory signals to aberrant kidney functions that allow sustained hypertension. By activating T lymphocytes, antigen-presenting cells such as dendritic cells represent an afferent sensing mechanism triggering T cell activation, cytokine generation, and renal salt and water reabsorption. Manipulating these inflammatory signals to attenuate hypertension without causing prohibitive systemic immunosuppression will pose a challenge, but disrupting actions of inflammatory mediators locally within the kidney may offer a path through which to target immune-mediated mechanisms of hypertension while capitalizing on Dr Dahl's key recognition of the kidney's importance in blood pressure regulation.
Collapse
Affiliation(s)
- Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, NC
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, NC
| |
Collapse
|
28
|
Xiao F, Farag MA, Xiao J, Yang X, Liu Y, Shen J, Lu B. The influence of phytochemicals on cell heterogeneity in chronic inflammation-associated diseases: the prospects of single cell sequencing. J Nutr Biochem 2022; 108:109091. [PMID: 35718097 DOI: 10.1016/j.jnutbio.2022.109091] [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: 11/08/2021] [Revised: 04/25/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
Abstract
Chronic inflammation-associated diseases include, but is not limited to cardiovascular disease, cancer, obesity, diabetes, etc. Cell heterogeneity is a prerequisite for understanding the physiological and pathological development of cell metabolism, and its response to external stimuli. Recently, dietary habits based on phytochemicals became increasingly recognized to play a pivotal role in chronic inflammation. Phytochemicals can relieve chronic inflammation by regulating inflammatory cell differentiation and immune cell response, but the influence of phytochemicals on cell heterogeneity from in vitro and ex vivo studies cannot simulate the complexity of cell differentiation in vivo due to the differences in cell lines and extracellular environment. Therefore, there is no consensus on the regulation mechanism of phytochemicals on chronic diseases based on cell heterogeneity. The purpose of this review is to summarize cell heterogeneity in common chronic inflammation-associated diseases and trace the effects of phytochemicals on cell differentiation in chronic diseases development. More importantly, by discussing the problems and challenges which hinder the study of cell heterogeneity in recent nutritional assessment experiments, we propose new prospects based on the drawbacks of existing research to optimize the research on the regulation mechanism of phytochemicals on chronic diseases. The need to explore precise measurements of cell heterogeneity is a key pillar in understanding the influence of phytochemicals on certain diseases. In the future, deeper understanding of cell-to-cell variation and the impact of food components and their metabolites on cell function by single-cell genomics and epigenomics with the focus on individual differences will open new avenues for the next generation of health care.
Collapse
Affiliation(s)
- Fan Xiao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini st., P.B. 11562, Cairo, Egypt; Department of Chemistry, School of Sciences & Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo-Ourense Campus, E-32004 Ourense, Spain
| | - Xuan Yang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Yan Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Jianfu Shen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Ningbo Research Institute, Zhejiang University, Ningbo, China.
| |
Collapse
|
29
|
Wan Z, Li X, Sun J, Li X, Liu Z, Dong H, Zhou Q, Qiu H, Xu J, Yang T, Wang WJ, Ou Y. Peripheral Blood Transcripts Predict Preoperative Obstructive Total Anomalous Pulmonary Venous Connection. Front Cardiovasc Med 2022; 9:892000. [PMID: 35711367 PMCID: PMC9194086 DOI: 10.3389/fcvm.2022.892000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of accessible noninvasive tools to examine the molecular alterations limits our understanding of the causes of total anomalous pulmonary venous connection (TAPVC), as well as the identification of effective operational strategies. Here, we consecutively enrolled peripheral leukocyte transcripts of 26 preoperative obstructive and 22 non-obstructive patients with TAPVC. Two-hundred and fifty six differentially expressed mRNA and 27 differentially expressed long noncoding RNA transcripts were dysregulated. The up-regulated mRNA was enriched in the hydrogen peroxide catabolic process, response to mechanical stimulus, neutrophil degranulation, hemostasis, response to bacterium, and the NABA CORE MATRISOME pathway, all of which are associated with the development of fibrosis. Furthermore, we constructed predictive models using multiple machine-learning algorithms and tested the performance in the validation set. The mRNA NR3C2 and lncRNA MEG3 were screened based on multiple iterations. The random forest prediction model can predict preoperative obstruction patients in the validation set with high accuracy (area under curve = 1; sensitivity = 1). These data highlight the potential of peripheral leukocyte transcripts to evaluate obstructive-related pathophysiological alterations, leading to precision healthcare solutions that could improve patient survival after surgery. It also provides a novel direction for the study of preoperative obstructive TAPVC.
Collapse
Affiliation(s)
- Zunmin Wan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohong Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jinghua Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Xiaohua Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Haojian Dong
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Hailong Qiu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Tingyu Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | | | - Yanqiu Ou
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Yanqiu Ou
| |
Collapse
|
30
|
Chyu KY, Zhao X, Zhou J, Dimayuga PC, Lio NW, Cercek B, Trac NT, Chung EJ, Shah PK. Immunization using ApoB-100 peptide-linked nanoparticles reduces atherosclerosis. JCI Insight 2022; 7:149741. [PMID: 35536648 PMCID: PMC9220835 DOI: 10.1172/jci.insight.149741] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Active immunization with the apolipoprotein B-100 (ApoB-100) peptide P210 reduces experimental atherosclerosis. To advance this immunization strategy to future clinical testing, we explored the possibility of delivering P210 as an antigen using nanoparticles, given this approach has been used clinically. We first characterized the responses of T cells to P210 using PBMCs from patients with atherosclerotic cardiovascular disease (ASCVD). We then investigated the use of P210 in self-assembling peptide amphiphile micelles (P210-PAMs) as a vaccine formulation to reduce atherosclerosis in B6.129P2-Apoetm1Unc/J (ApoE–/–) mice and P210’s potential mechanisms of action. We also generated and characterized a humanized mouse model with chimeric HLA-A*02:01/Kb in ApoE–/– background to test the efficacy of P210-PAM immunization as a bridge to future clinical testing. P210 provoked T cell activation and memory response in PBMCs of patients with ASCVD. Dendritic cell uptake of P210-PAM and its costaining with MHC-I molecules supported its use as a vaccine formulation. In ApoE–/– mice, immunization with P210-PAMs dampened P210-specific CD4+ T cell proliferative response and CD8+ T cell cytolytic response, modulated macrophage phenotype, and significantly reduced aortic atherosclerosis. Potential clinical relevance of P210-PAM immunization was demonstrated by reduced atherosclerosis in the humanized ApoE–/– mouse model. Our data support experimental and translational use of P210-PAM as a potential vaccine candidate against human ASCVD.
Collapse
Affiliation(s)
- Kuang-Yuh Chyu
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Xiaoning Zhao
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Jianchang Zhou
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Paul C Dimayuga
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Nicole Wm Lio
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Bojan Cercek
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| | - Noah T Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, United States of America
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, United States of America
| | - Prediman K Shah
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Heart Institute, Los Angeles, United States of America
| |
Collapse
|
31
|
Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol 2022; 22:251-265. [PMID: 34389841 PMCID: PMC10111155 DOI: 10.1038/s41577-021-00584-1] [Citation(s) in RCA: 233] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the root cause of many cardiovascular diseases. Extensive research in preclinical models and emerging evidence in humans have established the crucial roles of the innate and adaptive immune systems in driving atherosclerosis-associated chronic inflammation in arterial blood vessels. New techniques have highlighted the enormous heterogeneity of leukocyte subsets in the arterial wall that have pro-inflammatory or regulatory roles in atherogenesis. Understanding the homing and activation pathways of these immune cells, their disease-associated dynamics and their regulation by microbial and metabolic factors will be crucial for the development of clinical interventions for atherosclerosis, including potentially vaccination-based therapeutic strategies. Here, we review key molecular mechanisms of immune cell activation implicated in modulating atherogenesis and provide an update on the contributions of innate and adaptive immune cell subsets in atherosclerosis.
Collapse
Affiliation(s)
- Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
32
|
Metabolism in atherosclerotic plaques: immunoregulatory mechanisms in the arterial wall. Clin Sci (Lond) 2022; 136:435-454. [PMID: 35348183 PMCID: PMC8965849 DOI: 10.1042/cs20201293] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023]
Abstract
Over the last decade, there has been a growing interest to understand the link between metabolism and the immune response in the context of metabolic diseases but also beyond, giving then birth to a new field of research. Termed 'immunometabolism', this interdisciplinary field explores paradigms of both immunology and metabolism to provided unique insights into different disease pathogenic processes, and the identification of new potential therapeutic targets. Similar to other inflammatory conditions, the atherosclerotic inflammatory process in the artery has been associated with a local dysregulated metabolic response. Thus, recent studies show that metabolites are more than just fuels in their metabolic pathways, and they can act as modulators of vascular inflammation and atherosclerosis. In this review article, we describe the most common immunometabolic pathways characterised in innate and adaptive immune cells, and discuss how macrophages' and T cells' metabolism may influence phenotypic changes in the plaque. Moreover, we discuss the potential of targeting immunometabolism to prevent and treat cardiovascular diseases (CVDs).
Collapse
|
33
|
Ramji DP, Ismail A, Chen J, Alradi F, Al Alawi S. Survey of In Vitro Model Systems for Investigation of Key Cellular Processes Associated with Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:39-56. [PMID: 35237957 DOI: 10.1007/978-1-0716-1924-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Atherosclerosis progression is associated with a complex array of cellular processes in the arterial wall, including endothelial cell activation/dysfunction, chemokine-driven recruitment of immune cells, differentiation of monocytes to macrophages and their subsequent transformation into lipid laden foam cells, activation of inflammasome and pro-inflammatory signaling, and migration of smooth muscle cells from the media to the intima. The use of in vitro model systems has considerably advanced our understanding of these atherosclerosis-associated processes and they are also often used in drug discovery and other screening platforms. This chapter will describe key in vitro model systems employed frequently in atherosclerosis research.
Collapse
Affiliation(s)
- Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | - Alaa Ismail
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Jing Chen
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | - Fahad Alradi
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
34
|
Mauersberger C, Hinterdobler J, Schunkert H, Kessler T, Sager HB. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front Cardiovasc Med 2022; 8:813984. [PMID: 35087886 PMCID: PMC8787128 DOI: 10.3389/fcvm.2021.813984] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide and leukocyte recruitment is a key element of this phenomenon, thus allowing immune cells to enter the arterial wall. There, in concert with accumulating lipids, the invading leukocytes trigger a plethora of inflammatory responses which promote the influx of additional leukocytes and lead to the continued growth of atherosclerotic plaques. The recruitment process follows a precise scheme of tethering, rolling, firm arrest, crawling and transmigration and involves multiple cellular and subcellular players. This review aims to provide a comprehensive up-to-date insight into the process of leukocyte recruitment relevant to atherosclerosis, each from the perspective of endothelial cells, monocytes and macrophages, neutrophils, T lymphocytes and platelets. In addition, therapeutic options targeting leukocyte recruitment into atherosclerotic lesions-or potentially arising from the growing body of insights into its precise mechanisms-are highlighted.
Collapse
Affiliation(s)
- Carina Mauersberger
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
35
|
van Duijn J, de Jong MJM, Benne N, Leboux RJT, van Ooijen ME, Kruit N, Foks AC, Jiskoot W, Bot I, Kuiper J, Slütter B. Tc17 CD8+ T cells accumulate in murine atherosclerotic lesions, but do not contribute to early atherosclerosis development. Cardiovasc Res 2021; 117:2755-2766. [PMID: 33063097 PMCID: PMC8683708 DOI: 10.1093/cvr/cvaa286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/04/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
AIMS CD8+ T cells can differentiate into subpopulations that are characterized by a specific cytokine profile, such as the Tc17 population that produces interleukin-17. The role of this CD8+ T-cell subset in atherosclerosis remains elusive. In this study, we therefore investigated the contribution of Tc17 cells to the development of atherosclerosis. METHODS AND RESULTS Flow cytometry analysis of atherosclerotic lesions from apolipoprotein E-deficient mice revealed a pronounced increase in RORγt+CD8+ T cells compared to the spleen, indicating a lesion-specific increase in Tc17 cells. To study whether and how the Tc17 subset affects atherosclerosis, we performed an adoptive transfer of Tc17 cells or undifferentiated Tc0 cells into CD8-/- low-density lipoprotein receptor-deficient mice fed a Western-type diet. Using flow cytometry, we showed that Tc17 cells retained a high level of interleukin-17A production in vivo. Moreover, Tc17 cells produced lower levels of interferon-γ than their Tc0 counterparts. Analysis of the aortic root revealed that the transfer of Tc17 cells did not increase atherosclerotic lesion size, in contrast to Tc0-treated mice. CONCLUSION These findings demonstrate a lesion-localized increase in Tc17 cells in an atherosclerotic mouse model. Tc17 cells appeared to be non-atherogenic, in contrast to their Tc0 counterpart.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Differentiation
- Cells, Cultured
- Disease Models, Animal
- Interferon-gamma/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Maaike J M de Jong
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Naomi Benne
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Romain J T Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Marieke E van Ooijen
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Nicky Kruit
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| |
Collapse
|
36
|
Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol 2021; 18:824-837. [PMID: 34253911 DOI: 10.1038/s41569-021-00582-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
T cells are among the most common cell types present in atherosclerotic plaques and are increasingly being recognized as a central mediator in atherosclerosis development and progression. At the same time, triglycerides and fatty acids have re-emerged as crucial risk factors for atherosclerosis. Triglycerides and fatty acids are important components of the milieu to which the T cell is exposed from the circulation to the plaque, and increasing evidence shows that fatty acids influence T cell function. In this Review, we discuss the effects of fatty acids on four components of the T cell response - metabolism, activation, proliferation and polarization - and the influence of these changes on the pathogenesis of atherosclerosis. We also discuss how quiescent T cells can undergo a type of metabolic reprogramming induced by exposure to fatty acids in the circulation that influences the subsequent functions of T cells after activation, such as in atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands.
- Netherlands Heart Institute, Utrecht, Netherlands.
| |
Collapse
|
37
|
Grievink HW, Smit V, Verwilligen RAF, Bernabé Kleijn MNA, Smeets D, Binder CJ, Yagita H, Moerland M, Kuiper J, Bot I, Foks AC. Stimulation of the PD-1 Pathway Decreases Atherosclerotic Lesion Development in Ldlr Deficient Mice. Front Cardiovasc Med 2021; 8:740531. [PMID: 34790707 PMCID: PMC8591266 DOI: 10.3389/fcvm.2021.740531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Signaling through the coinhibitory programmed death (PD)-1/PD-L1 pathway regulates T cell responses and can inhibit ongoing immune responses. Inflammation is a key process in the development of atherosclerosis, the underlying cause for the majority of cardiovascular diseases. Dampening the excessive immune response that occurs during atherosclerosis progression by promoting PD-1/PD-L1 signaling may have a high therapeutic potential to limit disease burden. In this study we therefore aimed to assess whether an agonistic PD-1 antibody can diminish atherosclerosis development. Methods and Results: Ldlr−/− mice were fed a western-type diet (WTD) while receiving 100 μg of an agonistic PD-1 antibody or control vehicle twice a week. Stimulation of the PD-1 pathway delayed the WTD-induced monocyte increase in the circulation up to 3 weeks and reduced T cell activation and proliferation. CD4+ T cell numbers in the atherosclerotic plaque were reduced upon PD-1 treatment. More specifically, we observed a 23% decrease in atherogenic IFNγ-producing splenic CD4+ T cells and a 20% decrease in cytotoxic CD8+ T cells, whereas atheroprotective IL-10 producing CD4+ T cells were increased with 47%. Furthermore, we found an increase in regulatory B cells, B1 cells and associated atheroprotective circulating oxLDL-specific IgM levels in agonistic PD-1-treated mice. This dampened immune activation following agonistic PD-1 treatment resulted in reduced atherosclerosis development (p < 0.05). Conclusions: Our data show that stimulation of the coinhibitory PD-1 pathway inhibits atherosclerosis development by modulation of T- and B cell responses. These data support stimulation of coinhibitory pathways as a potential therapeutic strategy to combat atherosclerosis.
Collapse
Affiliation(s)
- Hendrika W Grievink
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands.,Centre for Human Drug Research, Leiden, Netherlands
| | - Virginia Smit
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Robin A F Verwilligen
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Mireia N A Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Diede Smeets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Hideo Yagita
- Department of Immunology, Juntendo University, Tokyo, Japan
| | - Matthijs Moerland
- Centre for Human Drug Research, Leiden, Netherlands.,Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| |
Collapse
|
38
|
Igel E, Haller A, Wolfkiel PR, Orr-Asman M, Jaeschke A, Hui DY. Distinct pro-inflammatory properties of myeloid cell-derived apolipoprotein E2 and E4 in atherosclerosis promotion. J Biol Chem 2021; 297:101106. [PMID: 34425108 PMCID: PMC8437825 DOI: 10.1016/j.jbc.2021.101106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 11/25/2022] Open
Abstract
Polymorphisms in the apolipoprotein E (apoE) gene are risk factors for chronic inflammatory diseases including atherosclerosis. The gene product apoE is synthesized in many cell types and has both lipid transport–dependent and lipid transport–independent functions. Previous studies have shown that apoE expression in myeloid cells protects against atherogenesis in hypercholesterolemic ApoE−/− mice. However, the mechanism of this protection is still unclear. Using human APOE gene replacement mice as models, this study showed that apoE2 and apoE4 expressed endogenously in myeloid cells enhanced the inflammatory response via mechanisms independent of plasma lipoprotein transport. The data revealed that apoE2-expressing myeloid cells contained higher intracellular cholesterol levels because of impaired efflux, causing increasing inflammasome activation and myelopoiesis. In contrast, intracellular cholesterol levels were not elevated in apoE4-expressing myeloid cells, and its proinflammatory property was found to be independent of inflammasome signaling and related to enhanced oxidative stress. When ApoE−/− mice were reconstituted with bone marrow from various human APOE gene replacement mice, effective reduction of atherosclerosis was observed with marrow cells obtained from APOE3 but not APOE2 and APOE4 gene replacement mice. Taken together, these results documented that apoE2 and apoE4 expression in myeloid cells promotes inflammation via distinct mechanisms and promotes atherosclerosis in a plasma lipoprotein transport–independent manner.
Collapse
Affiliation(s)
- Emily Igel
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - April Haller
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Patrick R Wolfkiel
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Melissa Orr-Asman
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
39
|
Muhammad K, Ayoub MA, Iratni R. Vascular Inflammation in Cardiovascular Disease: Is Immune System Protective or Bystander? Curr Pharm Des 2021; 27:2141-2150. [PMID: 33461451 DOI: 10.2174/1381612827666210118121952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/15/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Chronic atherosclerosis induced vascular inflammation and perturbation of lipid metabolism is believed to be a major cause of CVD. Interplay of innate and adaptive Immune system has been interwined with various risk factors associated with the initiation and progression of atherosclerosis in CVD. A large body of evidence indicates a correlation between immunity and atherosclerosis. Retention of plasma lipoproteins in arterial subendothelial wall triggers the T helper type 1 (Th1) cells and monocyte-derived macrophages to form atherosclerotic plaques. In the present review, we will discuss the pathogenesis of CVD in relation to atherosclerosis with a particular focus on pro-atherogenic role of immune cells. Recent findings have also suggested anti-atherogenic roles of different B cell subsets. Therapeutic approaches to target atherosclerosis risk factors have reduced the mortality, but a need exists for the novel therapies to treat arterial vascular inflammation. These insights into the immune pathogenesis of atherosclerosis can lead to new targeted therapeutics to abate cardiovascular mortality and morbidity.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed A Ayoub
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
40
|
Dounousi E, Duni A, Naka KK, Vartholomatos G, Zoccali C. The Innate Immune System and Cardiovascular Disease in ESKD: Monocytes and Natural Killer Cells. Curr Vasc Pharmacol 2021; 19:63-76. [PMID: 32600233 DOI: 10.2174/1570161118666200628024027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Adverse innate immune responses have been implicated in several disease processes, including cardiovascular disease (CVD) and chronic kidney disease (CKD). The monocyte subsets natural killer (NK) cells and natural killer T (NKT) cells are involved in innate immunity. Monocytes subsets are key in atherogenesis and the inflammatory cascade occurring in heart failure. Upregulated activity and counts of proinflammatory CD16+ monocyte subsets are associated with clinical indices of atherosclerosis, heart failure syndromes and CKD. Advanced CKD is a complex state of persistent systemic inflammation characterized by elevated expression of proinflammatory and pro-atherogenic CD14++CD16+ monocytes, which are associated with cardiovascular events and death both in the general population and among patients with CKD. Diminished NK cells and NKT cells counts and aberrant activity are observed in both coronary artery disease and end-stage kidney disease. However, evidence of the roles of NK cells and NKT cells in atherogenesis in advanced CKD is circumstantial and remains to be clarified. This review describes the available evidence regarding the roles of specific immune cell subsets in the pathogenesis of CVD in patients with CKD. Future research is expected to further uncover the links between CKD associated innate immune system dysregulation and accelerated CVD and will ideally be translated into therapeutic targets.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Anila Duni
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- 2nd Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece
| | - Carmine Zoccali
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| |
Collapse
|
41
|
Afonso MS, Sharma M, Schlegel M, van Solingen C, Koelwyn GJ, Shanley LC, Beckett L, Peled D, Rahman K, Giannarelli C, Li H, Brown EJ, Khodadadi-Jamayran A, Fisher EA, Moore KJ. miR-33 Silencing Reprograms the Immune Cell Landscape in Atherosclerotic Plaques. Circ Res 2021; 128:1122-1138. [PMID: 33593073 PMCID: PMC8049965 DOI: 10.1161/circresaha.120.317914] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Milessa Silva Afonso
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Monika Sharma
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Martin Schlegel
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
- Department of Anesthesiology and Intensive Care, Technical University of Munich School of Medicine, Germany (M. Schlegel)
| | - Coen van Solingen
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Graeme J Koelwyn
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Lianne C Shanley
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Lauren Beckett
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
| | - Daniel Peled
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Karishma Rahman
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Chiara Giannarelli
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY (C.G.)
| | - Huilin Li
- Division of Biostatics, Department of Population Health (H.L), New York University School of Medicine
| | - Emily J Brown
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | | | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Kathryn J Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| |
Collapse
|
42
|
Ait-Oufella H, Lavillegrand JR, Tedgui A. Regulatory T Cell-Enhancing Therapies to Treat Atherosclerosis. Cells 2021; 10:cells10040723. [PMID: 33805071 PMCID: PMC8064079 DOI: 10.3390/cells10040723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Experimental studies have provided strong evidence that chronic inflammation triggered by the sub-endothelial accumulation of cholesterol-rich lipoproteins in arteries is essential in the initiation and progression of atherosclerosis. Recent clinical trials highlighting the efficacy of anti-inflammatory therapies in coronary patients have confirmed that this is also true in humans Monocytes/macrophages are central cells in the atherosclerotic process, but adaptive immunity, through B and T lymphocytes, as well as dendritic cells, also modulates the progression of the disease. Analysis of the role of different T cell subpopulations in murine models of atherosclerosis identified effector Th1 cells as proatherogenic, whereas regulatory T cells (Tregs) have been shown to protect against atherosclerosis. For these reasons, better understanding of how Tregs influence the atherosclerotic process is believed to provide novel Treg-targeted therapies to combat atherosclerosis. This review article summarizes current knowledge about the role of Tregs in atherosclerosis and discusses ways to enhance their function as novel immunomodulatory therapeutic approaches against cardiovascular disease.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
- AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Saint-Antoine, Sorbonne Université, 75012 Paris, France
- Correspondence: ; Tel.: +33-1-5398-8006; Fax: +33-1-5398-8052
| | - Jean-Rémi Lavillegrand
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
| | - Alain Tedgui
- Paris Cardiovascular Research Center—PARCC, Université de Paris, INSERM UMR-S 970, 75012 Paris, France; (J.-R.L.); (A.T.)
| |
Collapse
|
43
|
Xia X, Wang M, Li J, Chen Q, Jin H, Liang X, Wang L. Identification of potential genes associated with immune cell infiltration in atherosclerosis. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2230-2242. [PMID: 33892542 DOI: 10.3934/mbe.2021112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
BACKGROUND This study aimed to analyze the potential genes associated with immune cell infiltration in atherosclerosis (AS). METHODS Gene expression profile data (GSE57691) of human arterial tissue samples were downloaded, and differentially expressed RNAs (DERNAs; long-noncoding RNA [lncRNAs], microRNAs [miRNAs], and messenger RNAs [mRNAs]) in AS vs. control groups were selected. Based on genome-wide expression levels, the proportion of infiltrating immune cells in each sample was assessed. Genes associated with immune infiltration were selected, and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Finally, a competing endogenous RNA (ceRNA) network was constructed, and the genes in the network were subjected to functional analyses. RESULTS A total of 1749 DERNAs meeting the thresholds were screened, including 1673 DEmRNAs, 63 DElncRNAs, and 13 DEmiRNAs. The proportions of B cells, CD4+ T cells, and CD8+ T cells were significantly different between the two groups. In total, 341 immune-associated genes such as HBB, FCN1, IL1B, CXCL8, RPS27A, CCN3, CTSZ, and SERPINA3 were obtained that were enriched in 70 significantly related GO biological processes (such as immune response) and 15 KEGG pathways (such as chemokine signaling pathway). A ceRNA network, including 33 lncRNAs, 11 miRNAs, and 216 mRNAs, was established. CONCLUSION Genes such as FCN1, IL1B, and SERPINA3 may be involved in immune cell infiltration and may play important roles in AS progression via ceRNA regulation.
Collapse
Affiliation(s)
- Xiaodong Xia
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin City, 300052, China
| | - Manman Wang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining City, 272000, Shandong, China
| | - Jiao Li
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin City, 300121, China
| | - Qiang Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin City, 300211, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin City, 300052, China
| | - Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin City, 300211, China
| | - Lijun Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin City, 300052, China
| |
Collapse
|
44
|
Potential Effects of Immunosuppression on Oxidative Stress and Atherosclerosis in Kidney Transplant Recipients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6660846. [PMID: 33688391 PMCID: PMC7920738 DOI: 10.1155/2021/6660846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/06/2021] [Accepted: 02/13/2021] [Indexed: 01/16/2023]
Abstract
Chronic kidney disease is a public health problem that, depending on the country, affects approximately 8-13% of the population, involving both males and females of all ages. Renal replacement therapy remains one of the most costly procedures. It is assumed that one of the factors influencing the course of chronic kidney disease might be oxidative stress. It is believed that the main mediators of oxidative stress are reactive oxygen species (ROS). Transiently increased concentrations of ROS play a significant role in maintaining an organism's homeostasis, as they are part of the redox-related signaling, and in the immune defense system, as they are produced in high amounts in inflammation. Systemic oxidative stress can significantly contribute to endothelial dysfunction along with exaggeration of atherosclerosis and development of cardiovascular disease, the leading cause of mortality in patients with kidney disease. Moreover, the progression of chronic kidney disease is strictly associated with the atherosclerotic process. Transplantation is the optimal method for renal replacement therapy. It improves better quality of life and prolongs survival compared with hemodialysis and peritoneal dialysis; however, even a successful transplantation does not correct the abnormalities found in chronic kidney disease. As transplantation reduces the concentration of uremic toxins, which are a factor of inflammation per se, both the procedure itself and the subsequent immunosuppressive treatment may be a factor that increases oxidative stress and hence vascular sclerosis and atherosclerotic cardiovascular disease. In the current work, we review the effect of several risk factors in kidney transplant recipients as well as immunosuppressive therapy on oxidative stress.
Collapse
|
45
|
Winkels H, Wolf D. Heterogeneity of T Cells in Atherosclerosis Defined by Single-Cell RNA-Sequencing and Cytometry by Time of Flight. Arterioscler Thromb Vasc Biol 2021; 41:549-563. [PMID: 33267666 PMCID: PMC7837690 DOI: 10.1161/atvbaha.120.312137] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022]
Abstract
The infiltration and accumulation of pro- and anti-inflammatory leukocytes within the intimal layer of the arterial wall is a hallmark of developing and progressing atherosclerosis. While traditionally perceived as macrophage- and foam cell-dominated disease, it is now established that atherosclerosis is a partial autoimmune disease that involves the recognition of peptides from ApoB (apolipoprotein B), the core protein of LDL (low-density lipoprotein) cholesterol particles, by CD4+ T-helper cells and autoantibodies against LDL and ApoB. Autoimmunity in the atherosclerotic plaque has long been understood as a pathogenic T-helper type-1 driven response with proinflammatory cytokine secretion. Recent developments in high-parametric cell immunophenotyping by mass cytometry, single-cell RNA-sequencing, and in tools exploring antigen-specificity have established the existence of several unforeseen layers of T-cell diversity with mixed TH1 and T regulatory cells transcriptional programs and unpredicted fates. These findings suggest that pathogenic ApoB-reactive T cells evolve from atheroprotective and immunosuppressive CD4+ T regulatory cells that lose their protective properties over time. Here, we discuss T-cell heterogeneity in atherosclerosis with a focus on plasticity, antigen-specificity, exhaustion, maturation, tissue residency, and its potential use in clinical prediction.
Collapse
Affiliation(s)
- Holger Winkels
- Department of Cardiology, Clinic III for Internal Medicine, University of Cologne, Germany. Department of Cardiology and Angiology I, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology, Clinic III for Internal Medicine, University of Cologne, Germany. Department of Cardiology and Angiology I, University Heart Center Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
46
|
Abstract
Atherosclerotic lesions are populated by cells of the innate and adaptive immune system, including CD8+ T cells. The CD8+ T cell infiltrate has recently been characterized in mouse and human atherosclerosis and revealed activated, cytotoxic, and possibly dysfunctional and exhausted cell phenotypes. In mouse models of atherosclerosis, antibody-mediated depletion of CD8+ T cells ameliorates atherosclerosis. CD8+ T cells control monopoiesis and macrophage accumulation in early atherosclerosis. In addition, CD8+ T cells exert cytotoxic functions in atherosclerotic plaques and contribute to macrophage cell death and necrotic core formation. CD8+ T cell activation may be antigen-specific, and epitopes of atherosclerosis-relevant antigens may be targets of CD8+ T cells and their cytotoxic activity. CD8+ T cell functions are tightly controlled by costimulatory and coinhibitory immune checkpoints. Subsets of regulatory CD25+CD8+ T cells with immunosuppressive functions can inhibit atherosclerosis. Importantly, local cytotoxic CD8+ T cell responses may trigger endothelial damage and plaque erosion in acute coronary syndromes. Understanding the complex role of CD8+ T cells in atherosclerosis may pave the way for defining novel treatment approaches in atherosclerosis. In this review article, we discuss these aspects, highlighting the emerging and critical role of CD8+ T cells in atherosclerosis.
Collapse
|
47
|
Mohmmad‐Rezaei M, Arefnezhad R, Ahmadi R, Abdollahpour‐Alitappeh M, Mirzaei Y, Arjmand M, Ferns GA, Bashash D, Bagheri N. An overview of the innate and adaptive immune system in atherosclerosis. IUBMB Life 2020. [DOI: 10.1002/iub.2425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mina Mohmmad‐Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | - Reza Arefnezhad
- Halal Research Center of IRI, FDA Tehran Iran
- Department of Anatomy, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | | | - Yousef Mirzaei
- Department of Biogeosciences, Scientific Research Center Soran University Soran Iraq
| | - Mohammad‐Hassan Arjmand
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
- Cancer Research Center Shahrekord University of Medical Sciences Shahrekord Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education Sussex United Kingdom
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| |
Collapse
|
48
|
Padgett LE, Dinh HQ, Wu R, Gaddis DE, Araujo DJ, Winkels H, Nguyen A, McNamara CA, Hedrick CC. Naive CD8 + T Cells Expressing CD95 Increase Human Cardiovascular Disease Severity. Arterioscler Thromb Vasc Biol 2020; 40:2845-2859. [PMID: 33054398 DOI: 10.1161/atvbaha.120.315106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) remains a significant global health concern with a high degree of mortality. While CD4+ T cells have been extensively studied in CVD, the importance of CD8+ T cells in this disease, despite their abundance and increased activation in human atherosclerotic plaques, remains largely unknown. Thus, the objective of this study was to compare peripheral T-cell signatures between humans with a high (severe) risk of CVD (including myocardial infarction or stroke) and those with a low risk of CVD. Approach and Results: Using mass cytometry, we uncovered a naive CD8+ T (TN) cell population expressing CD95 (termed CD95+CD8+ stem cell memory T [CD8 TSCM] cells) that was enriched in patients with high compared with low CVD. This T-cell subset enrichment within individuals with high CVD was a relative increase and resulted from the loss of CD95lo cells within the TN compartment. We found that CD8 TSCM cells positively correlated with CVD risk in humans, while CD8+ TN cells were inversely correlated. Atherosclerotic apolipoprotein E-deficient (ApoE-/-) mice also displayed respective 7- and 2-fold increases in CD8+ TSCM frequencies within the peripheral blood and aorta-draining paraaortic lymph nodes compared with C57BL/6J mice. CD8+ TSCM cells were 1.7-fold increased in aortas from western diet fed ApoE-/- mice compared with normal laboratory diet-fed ApoE-/- mice. Importantly, transfer of TSCM cells into immune-deficient Rag.Ldlr recipient mice that lacked T cells increased atherosclerosis, illustrating the importance of these cells in atherogenesis. CONCLUSIONS CD8+ TSCM cells are increased in humans with high CVD. As these TSCM cells promote atherosclerosis, targeting them may attenuate atherosclerotic plaque progression.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Huy Q Dinh
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Runpei Wu
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Dalia E Gaddis
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Daniel J Araujo
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Holger Winkels
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Anh Nguyen
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Coleen A McNamara
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| |
Collapse
|
49
|
Rai A, Narisawa M, Li P, Piao L, Li Y, Yang G, Cheng XW. Adaptive immune disorders in hypertension and heart failure: focusing on T-cell subset activation and clinical implications. J Hypertens 2020; 38:1878-1889. [PMID: 32890260 DOI: 10.1097/hjh.0000000000002456] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
: Hypertension is a growing health concern worldwide. Established hypertension is a causative factor of heart failure, which is characterized by increased vascular resistance and intractable uncontrolled blood pressure. Hypertension and heart failure have multiple causes and complex pathophysiology but cellular immunity is thought to contribute to the development of both. Recent studies showed that T cells play critical roles in hypertension and heart failure in humans and animals, with various stimuli leading to the formation of effector T cells that infiltrate the cardiovascular wall. Monocytes/macrophages also accumulate in the cardiovascular wall. Various cytokines (e.g. interleukin-6, interleukin-17, interleukin-10, tumor necrosis factor-α, and interferon-γ) released from immune cells of various subtypes promote vascular senescence and elastic laminal degradation as well as cardiac fibrosis and/or hypertrophy, leading to cardiovascular structural alterations and dysfunction. Recent laboratory evidence has defined a link between inflammation and the immune system in initiation and progression of hypertension and heart failure. Moreover, cross-talk among natural killer cells, adaptive immune cells (T cells and B cells), and innate immune cells (i.e. monocytes, macrophages, neutrophils, and dendritic cells) contributes to end-cardiovasculature damage and dysfunction in hypertension and heart failure. Clinical and experimental studies on the diagnostic potential of T-cell subsets revealed that blood regulatory T cells, CD4 cells, CD8 T cells, and the ratio of CD4 to CD8 T cells show promise as biomarkers of hypertension and heart failure. Therapeutic interventions to suppress activation of these cells may prove beneficial in reducing end-organ damage and preventing consequences of cardiovascular failure, including hypertension of heart failure.
Collapse
Affiliation(s)
- Avinas Rai
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Limei Piao
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Yanglong Li
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Guang Yang
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| |
Collapse
|
50
|
Seijkens TTP, Poels K, Meiler S, van Tiel CM, Kusters PJH, Reiche M, Atzler D, Winkels H, Tjwa M, Poelman H, Slütter B, Kuiper J, Gijbels M, Kuivenhoven JA, Matic LP, Paulsson-Berne G, Hedin U, Hansson GK, Nicolaes GAF, Daemen MJAP, Weber C, Gerdes N, de Winther MPJ, Lutgens E. Deficiency of the T cell regulator Casitas B-cell lymphoma-B aggravates atherosclerosis by inducing CD8+ T cell-mediated macrophage death. Eur Heart J 2020; 40:372-382. [PMID: 30452556 PMCID: PMC6340101 DOI: 10.1093/eurheartj/ehy714] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
Aims The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb−/−Apoe−/− mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb−/−Apoe−/− macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb−/−Apoe−/− CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb−/−Apoe−/− bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques. ![]()
Collapse
Affiliation(s)
- Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Svenja Meiler
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Myrthe Reiche
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Goethestraße 33D, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Holger Winkels
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Marc Tjwa
- Laboratory of Vascular Hematology/Angiogenesis, Institute for Transfusion Medicine, Goethe University Frankfurt, Sandhofstraße 1D, Germany
| | - Hessel Poelman
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER, Maastricht University, Maastricht, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einstein weg 55, 2333 CC, Leiden, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einstein weg 55, 2333 CC, Leiden, the Netherlands
| | - Marion Gijbels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center Groningen, Postbus 72, AB Groningen, The Netherlands
| | - Ljubica Perisic Matic
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Gabrielle Paulsson-Berne
- Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Solna SE-171 76 Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Göran K Hansson
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Solna, SE-171 76, Stockholm, Sweden
| | - Gerry A F Nicolaes
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER, Maastricht University, Maastricht, the Netherlands
| | - Mat J A P Daemen
- Department of Pathology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany.,Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Moorenstraße 5m 0225 Düsseldorf, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Room K1-110, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Pettenkoferstraße 8a & 9, Munich, Germany
| |
Collapse
|