1
|
de Jong MJM, Depuydt MAC, Schaftenaar FH, Liu K, Maters D, Wezel A, Smeets HJ, Kuiper J, Bot I, van Gisbergen K, Slütter B. Resident Memory T Cells in the Atherosclerotic Lesion Associate With Reduced Macrophage Content and Increased Lesion Stability. Arterioscler Thromb Vasc Biol 2024; 44:1318-1329. [PMID: 38634281 DOI: 10.1161/atvbaha.123.320511] [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: 11/29/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Tissue resident memory T (TRM) cells are a T-cell subset that resides at the site of prior antigen recognition to protect the body against reoccurring encounters. Besides their protective function, TRM cells have also been implicated in inflammatory disorders. TRM cells are characterized by the expression of CD69 and transcription factors Hobit (homolog of Blimp-1 [B lymphocyte-induced maturation protein 1] in T cells) and Blimp-1. As the majority of T cells in the arterial intima expresses CD69, TRM cells may contribute to the pathogenesis of atherosclerosis as well. Here, we aimed to assess the presence and potential role of TRM cells in atherosclerosis. METHODS To identify TRM cells in human atherosclerotic lesions, a single-cell RNA-sequencing data set was interrogated, and T-cell phenotypes were compared with that of integrated predefined TRM cells. The presence and phenotype of TRM in atherosclerotic lesions was corroborated using a mouse model that enabled tracking of Hobit-expressing TRM cells. To explore the function of TRM cells during atherogenesis, RAG1-/- (recombination activating gene 1 deficient) LDLr-/- (low-density lipoprotein receptor knockout) mice received a bone marrow transplant from HobitKO/CREBlimp-1flox/flox mice, which exhibit abrogated TRM cell formation, whereafter the mice were fed a Western-type diet for 10 weeks. RESULTS Human atherosclerotic lesions contained T cells that exhibited a TRM cell-associated gene signature. Moreover, a fraction of these T cells clustered together with predefined TRM cells upon integration. The presence of Hobit-expressing TRM cells in the atherosclerotic lesion was confirmed in mice. These lesion-derived TRM cells were characterized by the expression of CD69 and CD49α. Moreover, we demonstrated that this small T-cell subset significantly affects lesion composition, by reducing the amount of intralesional macrophages and increasing collagen content. CONCLUSIONS TRM cells, characterized by the expression of CD69 and CD49α, constitute a minor population in atherosclerotic lesions and are associated with increased lesion stability in a Hobit and Blimp-1 knockout mouse model.
Collapse
MESH Headings
- Animals
- Atherosclerosis/pathology
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Humans
- Memory T Cells/immunology
- Memory T Cells/metabolism
- Macrophages/metabolism
- Macrophages/immunology
- Macrophages/pathology
- Disease Models, Animal
- Immunologic Memory
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Plaque, Atherosclerotic
- Mice, Inbred C57BL
- Mice
- Male
- Mice, Knockout
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Phenotype
- Female
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Aortic Diseases/pathology
- Aortic Diseases/immunology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
Collapse
Affiliation(s)
- Maaike J M de Jong
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Marie A C Depuydt
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Frank H Schaftenaar
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Kun Liu
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - David Maters
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center, The Hague, the Netherlands (A.W., H.J.S.)
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center, The Hague, the Netherlands (A.W., H.J.S.)
| | - Johan Kuiper
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Ilze Bot
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| | - Klaas van Gisbergen
- van Gisbergen Lab, Tissue Immunity, Champalimaud Research, Lisbon, Portugal (K.v.G.)
| | - Bram Slütter
- Division of Biotherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands (M.J.M.d.J., M.A.C.D., F.H.S., K.L., D.M., J.K., I.B., B.S.)
| |
Collapse
|
2
|
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
|
3
|
Shi X, Qu M, Jiang Y, Zhu Z, Dai C, Jiang M, Ding L, Yan Y, Wang C, Zhang X, Cheng S, Hao X. Association of immune cell composition with the risk factors and incidence of acute coronary syndrome. Clin Epigenetics 2023; 15:115. [PMID: 37461090 PMCID: PMC10353119 DOI: 10.1186/s13148-023-01527-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Although immune cells are involved in acute coronary syndrome (ACS), few studies have explored the association of incident ACS with the relative immune cell proportions. We aimed to investigate the association of immune cell proportions with the incidence and risk factors of ACS in the Dongfeng-Tongji cohort. METHODS We conducted the analyses with 38,295 subjects from the first follow-up of the Dongfeng-Tongji cohort, including DNA methylation profiles for 1570 individuals. The proportions of immune cell types were observed from routine blood tests or estimated from DNA methylation profiles. For both observed and estimated immune cell proportions, we tested their associations with risk factors of ACS by multivariable linear regression models. In addition, the association of each immune cell proportion with incident ACS was assessed by the Cox regression model and conditional logistic regression model, respectively, adjusting for the risk factors of ACS. FINDINGS The proportions of lymphocytes, monocytes, and neutrophils showed strong associations with sex, followed by diabetes. Moreover, sex and current smoking were the two factors with strongest association with the proportions of lymphocyte subtypes. The hazard ratio (HR) and 95% confidence interval (CI) of incident ACS per standard deviation (SD) increase in proportions of lymphocytes and neutrophils were 0.91 (0.85-0.96) and 1.10 (1.03-1.16), respectively. Furthermore, the OR (95% CI) of incident ACS per SD increase in proportions of NK cells, CD4+ T cells, and B cells were 0.88 (0.78-0.99), 1.15 (1.03-1.30), and 1.13 (1.00-1.26), respectively. INTERPRETATION The proportions of immune cells were associated with several risk factors of ACS, including sex, diabetes, and current smoking. In addition, proportion of neutrophils had a risk effect, while proportion of lymphocytes had a protective effect on the incidence of ACS. The protective effect of lymphocytes was probably driven by NK cells.
Collapse
Affiliation(s)
- Xian Shi
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghan Qu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Jiang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Zhu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengguqiu Dai
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghui Jiang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Ding
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
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
|
5
|
Su C, Lu Y, Wang Z, Guo J, Hou Y, Wang X, Qin Z, Gao J, Sun Z, Dai Y, Liu Y, Liu G, Xian X, Cui X, Zhang J, Tang J. Atherosclerosis: The Involvement of Immunity, Cytokines and Cells in Pathogenesis, and Potential Novel Therapeutics. Aging Dis 2022:AD.2022.1208. [PMID: 37163428 PMCID: PMC10389830 DOI: 10.14336/ad.2022.1208] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/08/2022] [Indexed: 05/12/2023] Open
Abstract
As a leading contributor to coronary artery disease (CAD) and stroke, atherosclerosis has become one of the major cardiovascular diseases (CVD) negatively impacting patients worldwide. The endothelial injury is considered to be the initial step of the development of atherosclerosis, resulting in immune cell migration and activation as well as inflammatory factor secretion, which further leads to acute and chronic inflammation. In addition, the inflammation and lipid accumulation at the lesions stimulate specific responses from different types of cells, contributing to the pathological progression of atherosclerosis. As a result, recent studies have focused on using molecular biological approaches such as gene editing and nanotechnology to mediate cellular response during atherosclerotic development for therapeutic purposes. In this review, we systematically discuss inflammatory pathogenesis during the development of atherosclerosis from a cellular level with a focus on the blood cells, including all types of immune cells, together with crucial cells within the blood vessel, such as smooth muscle cells and endothelial cells. In addition, the latest progression of molecular-cellular based therapy for atherosclerosis is also discussed. We hope this review article could be beneficial for the clinical management of atherosclerosis.
Collapse
Affiliation(s)
- Chang Su
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yongzheng Lu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zeyu Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiacheng Guo
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yachen Hou
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Xiaofang Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhen Qin
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiamin Gao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhaowei Sun
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yichen Dai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yu Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Guozhen Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, Peking University, Beijing, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Jinying Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Junnan Tang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Roberts LB, Lord GM, Howard JK. Heartbreakers or Healers? Innate Lymphoid Cells in Cardiovascular Disease and Obesity. Front Immunol 2022; 13:903678. [PMID: 35634348 PMCID: PMC9130471 DOI: 10.3389/fimmu.2022.903678] [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: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are responsible for most pre-mature deaths worldwide, contributing significantly to the global burden of disease and its associated costs to individuals and healthcare systems. Obesity and associated metabolic inflammation underlie development of several major health conditions which act as direct risk factors for development of CVDs. Immune system responses contribute greatly to CVD development and progression, as well as disease resolution. Innate lymphoid cells (ILCs) are a family of helper-like and cytotoxic lymphocytes, typically enriched at barrier sites such as the skin, lung, and gastrointestinal tract. However, recent studies indicate that most solid organs and tissues are home to resident populations of ILCs - including those of the cardiovascular system. Despite their relative rarity, ILCs contribute to many important biological effects during health, whilst promoting inflammatory responses during tissue damage and disease. This mini review will discuss the evidence for pathological and protective roles of ILCs in CVD, and its associated risk factor, obesity.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| |
Collapse
|
7
|
Analysis of Immune and Inflammation Characteristics of Atherosclerosis from Different Sample Sources. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5491038. [PMID: 35509837 PMCID: PMC9060985 DOI: 10.1155/2022/5491038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/21/2022] [Indexed: 11/17/2022]
Abstract
Background Atherosclerosis is the predominant cause of cardiovascular diseases. Existing studies suggest that the development of atherosclerosis is closely related to inflammation and immunity, but whether there are differences and similarities between atherosclerosis occurring at different sites is still unknown. We elucidated the pathological characteristics of peripheral vascular diseases by using bioinformatic analyses on immune cells and inflammation-related gene expression in atherosclerotic arteries and plaques. Methods Eight data sets regarding atherosclerosis were downloaded from the Gene Expression Omnibus database. Human immune genes were obtained from the IMMPORT website. The samples were scored and divided into high- and low-immune groups. Then the samples were analysed using weighted gene co-expression network analysis, while the modules were analysed using functional enrichment. The protein–protein interaction network was constructed using the STRING and Cytoscape databases. The hub immune genes were screened, and the correlation between hub immune genes and immune cells was analysed. Results Immune cells and their functions were significantly different during atherosclerosis development. The infiltration proportion of immune cells was approximately similar in samples from different sources of patients with carotid atherosclerosis. However, the sensitivity of lower extremity atherosclerosis samples to immune cells is lower than that of carotid atherosclerosis samples.The samples from the plaque and artery were mainly infiltrated by macrophages, T cells and mast cells. After immune cells were assessed, resting NK cells, activated mast cells and M0 macrophages were found to be key immune cells in atherosclerosis and plaque formation. In addition, CCL4, TLR2, IL1B and PTPRC were considered to be immune marker genes in atherosclerosis development. Conclusion. Bioinformatic data analysis confirms the essential role of immune cells in cardiovascular diseases, and also indicates some differences of immune and inflammation characteristics of atherosclerosis between carotid and lower extremity arteries.
Collapse
|
8
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 475] [Impact Index Per Article: 158.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
9
|
Nour J, Moregola A, Molgora M, Mantovani A, Uboldi P, Catapano A, Garlanda C, Bonacina F, Norata GD. Interleukin 1 receptor 8 deficiency does not impact atherosclerosis. Thromb Haemost 2022; 122:1833-1836. [PMID: 35436796 DOI: 10.1055/a-1827-7205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
No abstract.
Collapse
Affiliation(s)
- Jasmine Nour
- Department of Excellence of Pharmacological and Biomolecular sciences, University of Milan, Milano, Italy
| | - Annalisa Moregola
- Department of Excellence of Pharmacological and Biomolecular sciences, University of Milan, Milano, Italy
| | | | - Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,William Harvey Research Institute, Queen Mary University of London, London, United Kingdom of Great Britain and Northern Ireland.,Humanitas University, Milan, Italy
| | | | - Alberico Catapano
- IRCCS MultiMedica, Sesto San Giovanni, Italy.,University of Milan, Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Fabrizia Bonacina
- Pharmacological and Biomolecular sciences, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological Sciences, University of Milan, Milan, Italy.,Centro SISA per lo Studio dell'Aterosclerosi, Hospital Bassini, Cinisello Balsamo, Italy
| |
Collapse
|
10
|
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: 272] [Impact Index Per Article: 90.7] [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
|
11
|
Murphy JM, Ngai L, Mortha A, Crome SQ. Tissue-Dependent Adaptations and Functions of Innate Lymphoid Cells. Front Immunol 2022; 13:836999. [PMID: 35359972 PMCID: PMC8960279 DOI: 10.3389/fimmu.2022.836999] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-resident immune cells reside in distinct niches across organs, where they contribute to tissue homeostasis and rapidly respond to perturbations in the local microenvironment. Innate lymphoid cells (ILCs) are a family of innate immune cells that regulate immune and tissue homeostasis. Across anatomical locations throughout the body, ILCs adopt tissue-specific fates, differing from circulating ILC populations. Adaptations of ILCs to microenvironmental changes have been documented in several inflammatory contexts, including obesity, asthma, and inflammatory bowel disease. While our understanding of ILC functions within tissues have predominantly been based on mouse studies, development of advanced single cell platforms to study tissue-resident ILCs in humans and emerging patient-based data is providing new insights into this lymphocyte family. Within this review, we discuss current concepts of ILC fate and function, exploring tissue-specific functions of ILCs and their contribution to health and disease across organ systems.
Collapse
Affiliation(s)
- Julia M. Murphy
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
12
|
Eberhardt N, Giannarelli C. How Single-Cell Technologies Have Provided New Insights Into Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:243-252. [PMID: 35109673 PMCID: PMC8966900 DOI: 10.1161/atvbaha.121.315849] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The development of innovative single-cell technologies has allowed the high-dimensional transcriptomic and proteomic profiling of individual blood and tissue cells. Recent single-cell studies revealed a new cellular heterogeneity of atherosclerotic plaque tissue and allowed a better understanding of distinct immune functional states in the context of atherosclerosis. In this brief review, we describe how single-cell technologies have shed a new light on the cellular composition of atherosclerotic plaques, and their response to diet perturbations or genetic manipulation in mouse models of atherosclerosis. We discuss how single-cell RNA sequencing, cellular indexing of transcriptomes and epitopes by sequencing, transposase-accessible chromatin with high-throughput sequencing, and cytometry by time-of-flight platforms have empowered the identification of discrete immune, endothelial, and smooth muscle cell alterations in atherosclerosis progression and regression. Finally, we review how single-cell approaches have allowed mapping the cellular and molecular composition of human atherosclerotic plaques and the discovery of new immune alterations in plaques from patients with stroke.
Collapse
Affiliation(s)
- Natalia Eberhardt
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,NYU Cardiovascular Research Center, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA
| | - Chiara Giannarelli
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,NYU Cardiovascular Research Center, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,Department of Pathology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,Correspondence to: Chiara Giannarelli, MD, PhD, 435 East 30th street, Science Building, New York, NY, 10016,
| |
Collapse
|
13
|
Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, Spinetti G, Ambrosio G, Bruno A. When a Friend Becomes Your Enemy: Natural Killer Cells in Atherosclerosis and Atherosclerosis-Associated Risk Factors. Front Immunol 2022; 12:798155. [PMID: 35095876 PMCID: PMC8793801 DOI: 10.3389/fimmu.2021.798155] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis (ATS), the change in structure and function of arteries with associated lesion formation and altered blood flow, is the leading cause of cardiovascular disease, the number one killer worldwide. Beyond dyslipidemia, chronic inflammation, together with aberrant phenotype and function of cells of both the innate and adaptive immune system, are now recognized as relevant contributors to atherosclerosis onset and progression. While the role of macrophages and T cells in atherosclerosis has been addressed in several studies, Natural Killer cells (NKs) represent a poorly explored immune cell type, that deserves attention, due to NKs’ emerging contribution to vascular homeostasis. Furthermore, the possibility to re-polarize the immune system has emerged as a relevant tool to design new therapies, with some succesfull exmples in the field of cancer immunotherapy. Thus, a deeper knowledge of NK cell pathophysiology in the context of atherosclerosis and atherosclerosis-associated risk factors could help developing new preventive and treatment strategies, and decipher the complex scenario/history from “the risk factors for atherosclerosis” Here, we review the current knowledge about NK cell phenotype and activities in atherosclerosis and selected atherosclerosis risk factors, namely type-2 diabetes and obesity, and discuss the related NK-cell oriented environmental signals.
Collapse
Affiliation(s)
- Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | - Martina Cucchiara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Federica Riccio
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gian Franco Gensini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | - Gaia Spinetti
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | | | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| |
Collapse
|
14
|
Gerhardt T, Haghikia A, Stapmanns P, Leistner DM. Immune Mechanisms of Plaque Instability. Front Cardiovasc Med 2022; 8:797046. [PMID: 35087883 PMCID: PMC8787133 DOI: 10.3389/fcvm.2021.797046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation crucially drives atherosclerosis from disease initiation to the emergence of clinical complications. Targeting pivotal inflammatory pathways without compromising the host defense could compliment therapy with lipid-lowering agents, anti-hypertensive treatment, and lifestyle interventions to address the substantial residual cardiovascular risk that remains beyond classical risk factor control. Detailed understanding of the intricate immune mechanisms that propel plaque instability and disruption is indispensable for the development of novel therapeutic concepts. In this review, we provide an overview on the role of key immune cells in plaque inception and progression, and discuss recently identified maladaptive immune phenomena that contribute to plaque destabilization, including epigenetically programmed trained immunity in myeloid cells, pathogenic conversion of autoreactive regulatory T-cells and expansion of altered leukocytes due to clonal hematopoiesis. From a more global perspective, the article discusses how systemic crises such as acute mental stress or infection abruptly raise plaque vulnerability and summarizes recent advances in understanding the increased cardiovascular risk associated with COVID-19 disease. Stepping outside the box, we highlight the role of gut dysbiosis in atherosclerosis progression and plaque vulnerability. The emerging differential role of the immune system in plaque rupture and plaque erosion as well as the limitations of animal models in studying plaque disruption are reviewed.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philip Stapmanns
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
| | - David Manuel Leistner
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: David Manuel Leistner
| |
Collapse
|
15
|
Abstract
Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.
Collapse
Affiliation(s)
- Dawn M Fernandez
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- New York University Cardiovascular Research Center, New York University Langone Health, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
16
|
Reevaluation of NOD/SCID Mice as NK Cell-Deficient Models. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8851986. [PMID: 34805408 PMCID: PMC8598338 DOI: 10.1155/2021/8851986] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 07/21/2021] [Accepted: 10/21/2021] [Indexed: 12/02/2022]
Abstract
Objective Natural killer (NK) cell-deficient mice are useful models in biomedical research. NOD/SCID mice have been used as a model of this type in research. However, the actual status of NK cells in NOD/SCID mice and CB17/SCID mice in comparison with that in BALB/c mice has not been sufficiently evaluated. Methods Splenocytes from naïve or poly(I:C)-treated mice were isolated for phenotyping and analysis of cytotoxicity-related molecules and inhibitory receptors; for cytotoxicity assay, purified NK cells were also used. Results The proportion of splenic NK cells did not differ significantly between NOD/SCID and CB17/SCID mice. The perforin levels in NK cells were similar between the poly(I:C)-treated CB17/SCID and NOD/SCID mice, while the granzyme B and NKG2A/C/E levels in NK cells from NOD/SCID mice were significantly lower than those from CB17/SCID mice. Moreover, the NKG2D and Ly49A levels in NK cells from NOD/SCID mice were higher than those from CB17/SCID. The splenocytes from CB17/SCID mice showed higher cytotoxicity than those from NOD/SCID mice, while the cytotoxicity of purified NK cells basically did not differ between the two strains. After in vitro stimulation with cytokines, the splenocytes from CB17/SCID mice showed higher IFN-γ production than those from NOD/SCID mice; however, NK cells did not. Conclusion There was no significant difference in the proportion of splenic NK cells between CB17/SCID and NOD/SCID mice, and the function of NK cells was only partially compromised in NOD/SCID mice. Caution should be taken when considering the use of NOD/SCID mice as an NK-deficient model.
Collapse
|
17
|
Aghamajidi A, Gorgani M, Shahba F, Shafaghat Z, Mojtabavi N. The potential targets in immunotherapy of atherosclerosis. Int Rev Immunol 2021; 42:199-216. [PMID: 34779341 DOI: 10.1080/08830185.2021.1988591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Cardiovascular disease is the most common cause of death, which has the highest mortality rate worldwide. Although a diverse range of inflammatory diseases can affect the cardiovascular system, however, heart failure and stroke occur due to atherosclerosis. Atherosclerosis is a chronic autoinflammatory disease of small to large vessels in which different immune mediators are involved in lipid plaque formation and inflammatory vascular remodeling process. A better understanding of the pathophysiology of atherosclerosis may lead to uncovering immunomodulatory therapies. Despite present diagnostic and therapeutic methods, the lack of immunotherapy in the prevention and treatment of atherosclerosis is perceptible. In this review, we will discuss the promising immunological-based therapeutics and novel preventive approaches for atherosclerosis. This study could provide new insights into a better perception of targeted therapeutic pathways and biological therapies.
Collapse
Affiliation(s)
- Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Melika Gorgani
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Faezeh Shahba
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Duni A, Vartholomatos G, Balafa O, Ikonomou M, Tseke P, Lakkas L, Rapsomanikis KP, Kitsos A, Theodorou I, Pappas C, Naka KK, Mitsis M, Dounousi E. The Association of Circulating CD14++CD16+ Monocytes, Natural Killer Cells and Regulatory T Cells Subpopulations With Phenotypes of Cardiovascular Disease in a Cohort of Peritoneal Dialysis Patients. Front Med (Lausanne) 2021; 8:724316. [PMID: 34746172 PMCID: PMC8565661 DOI: 10.3389/fmed.2021.724316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
The altered expression of immune cells including monocyte subsets, natural killer (NK) cells and CD4+CD25+ regulatory T cells (Tregs) in end-stage kidney disease, affect the modulation of inflammation and immunity with significant clinical implications. The aim of this study was to investigate the profile of specific immune cells subpopulations and their correlations with phenotypes of established cardiovascular disease (CVD), including coronary artery disease (CAD) and heart failure (HF) in peritoneal dialysis (PD) patients. Materials and Methods: 29 stable PD patients and 13 healthy volunteers were enrolled. Demographic, laboratory, bioimpedance measurements, lung ultrasound and echocardiography data were collected. The peripheral blood immune cell subsets analysis was performed using flow cytometry. Results: PD patients compared to normal controls had lower total lymphocytes (22.3 ± 6.28 vs. 31.3 ± 5.54%, p = <0.001) and B-lymphocytes (6.39 ± 3.75 vs. 9.72 ± 3.63%, p = 0.01) as well as higher CD14++CD16+ monocytes numbers (9.28 ± 6.36 vs. 4.75 ± 2.75%, p = 0.0002). PD patients with prevalent CAD had NK cells levels elevated above median values (85.7 vs. 40.9%, p = 0.04) and lower B cells counts (3.85 ± 2.46 vs. 7.2 ± 3.77%, p = 0.03). Patients with increased NK cells (>15.4%) had 3.8 times higher risk of CAD comparing with patients with lower NK cell levels (95% CI, 1.86 – 77.87; p = 0.034). B cells were inversely associated with the presence of CAD (increase of B-lymphocyte by 1% was associated with 30% less risk for presence of CAD (95% CI, −0.71 – 0.01; p = 0.05). Overhydrated patients had lower lymphocytes counts (18.3 ± 4.29% vs. 24.7 ± 6.18%, p = 0.006) and increased NK cells [20.5% (14.3, 23.6) vs. 13.21% (6.23, 19.2), p = 0.04)]. In multiple logistic regression analysis the CRP (OR 1.43; 95% CI, 1.00 – 2.05; p = 0.04)] and lymphocytes counts (OR 0.79; 95% CI, 0.63–0.99; p = 0.04)] were associated with the presence of lung comets. Patients with higher NK cells (>15.4%, n = 15) were more likely to be rapid transporters (D/P creatinine 0.76 ± 0.1 vs. 0.69 ± 0.08, p = 0.04). Patients displaying higher Tregs (>1.79%) were older (70.8 ± 10.7 years vs. 57.7 ± 14.7years, p = 0.011) and had higher nPCR (0.83 ± 0.14 vs. 0.91 ± 0.17, p = 0.09). Conclusion: Future research is required to evaluate the role of immune cells subsets as potential tools to identify patients at the highest risk for complications and guide interventions.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece
| | - Olga Balafa
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Margarita Ikonomou
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | | | - Lampros Lakkas
- Second Department of Cardiology and Michaelidion Cardiac Center, Medical School University of Ioannina, Ioannina, Greece
| | | | - Athanasios Kitsos
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Ioanna Theodorou
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Charalambos Pappas
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology and Michaelidion Cardiac Center, Medical School University of Ioannina, Ioannina, Greece
| | - Michael Mitsis
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece.,Department of Surgery, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece.,Department of Nephrology, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
19
|
Ghilas S, Ambrosini M, Cancel JC, Brousse C, Massé M, Lelouard H, Dalod M, Crozat K. Natural killer cells and dendritic epidermal γδ T cells orchestrate type 1 conventional DC spatiotemporal repositioning toward CD8 + T cells. iScience 2021; 24:103059. [PMID: 34568787 PMCID: PMC8449251 DOI: 10.1016/j.isci.2021.103059] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/14/2021] [Accepted: 08/25/2021] [Indexed: 02/03/2023] Open
Abstract
Successful immune responses rely on a regulated delivery of the right signals to the right cells at the right time. Here we show that natural killer (NK) and dendritic epidermal γδ T cells (DETCs) use similar mechanisms to spatiotemporally orchestrate conventional type 1 dendritic cell (cDC1) functions in the spleen, skin, and its draining lymph nodes (dLNs). Upon MCMV infection in the spleen, cDC1 clusterize with activated NK cells in marginal zones. This XCR1-dependent repositioning of cDC1 toward NK cells allows contact delivery of IL-12 and IL-15/IL-15Rα by cDC1, which is critical for NK cell responses. NK cells deliver granulocyte-macrophage colony-stimulating factor (GM-CSF) to cDC1, guiding their CCR7-dependent relocalization into the T cell zone. In MCMV-infected skin, XCL1-secreting DETCs promote cDC1 migration from the skin to the dLNs. This XCR1-dependent licensing of cDC1 both in the spleen and skin accelerates antiviral CD8+ T cell responses, revealing an additional mechanism through which cDC1 bridge innate and adaptive immunity. Upon viral infection in the spleen, NK cells clusterize with cDC1 in the marginal zone This XCL1/XCR1-dependent interaction allows mutual delivery of activating signals NK cell GM-CSF directs cDC1 migration to T cell zone boosting CD8+ T cell priming In the skin, DETCs contact cDC1 via XCL1/XCR1 to promote antiviral CD8+ T cell priming
Collapse
Affiliation(s)
- Sonia Ghilas
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Ambrosini
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Jean-Charles Cancel
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Carine Brousse
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marion Massé
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Hugues Lelouard
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Dalod
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Karine Crozat
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| |
Collapse
|
20
|
Xing H, Pang H, Du T, Yang X, Zhang J, Li M, Zhang S. Establishing a Risk Prediction Model for Atherosclerosis in Systemic Lupus Erythematosus. Front Immunol 2021; 12:622216. [PMID: 33936038 PMCID: PMC8085548 DOI: 10.3389/fimmu.2021.622216] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/19/2021] [Indexed: 12/21/2022] Open
Abstract
Background and aims: Patients with systemic lupus erythematosus (SLE) have a significantly higher incidence of atherosclerosis than the general population. Studies on atherosclerosis prediction models specific for SLE patients are very limited. This study aimed to build a risk prediction model for atherosclerosis in SLE. Methods: RNA sequencing was performed on 67 SLE patients. Subsequently, differential expression analysis was carried out on 19 pairs of age-matched SLE patients with (AT group) or without (Non-AT group) atherosclerosis using peripheral venous blood. We used logistic least absolute shrinkage and selection operator regression to select variables among differentially expressed (DE) genes and clinical features and utilized backward stepwise logistic regression to build an atherosclerosis risk prediction model with all 67 patients. The performance of the prediction model was evaluated by area under the curve (AUC), calibration curve, and decision curve analyses. Results: The 67 patients had a median age of 42.7 (Q1-Q3: 36.6-51.2) years, and 20 (29.9%) had atherosclerosis. A total of 106 DE genes were identified between the age-matched AT and Non-AT groups. Pathway analyses revealed that the AT group had upregulated atherosclerosis signaling, oxidative phosphorylation, and interleukin (IL)-17-related pathways but downregulated T cell and B cell receptor signaling. Keratin 10, age, and hyperlipidemia were selected as variables for the risk prediction model. The AUC and Hosmer-Lemeshow test p-value of the model were 0.922 and 0.666, respectively, suggesting a relatively high discrimination and calibration performance. The prediction model had a higher net benefit in the decision curve analysis than that when predicting with age or hyperlipidemia only. Conclusions: We built an atherosclerotic risk prediction model with one gene and two clinical factors. This model may greatly assist clinicians to identify SLE patients with atherosclerosis, especially asymptomatic atherosclerosis.
Collapse
Affiliation(s)
- Haiping Xing
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Haiyu Pang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Tian Du
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China.,Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xufei Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Zhang
- Department of Diagnostic Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Mengtao Li
- Key Laboratory of Rheumatology and Clinical Immunology, Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Ministry of Education, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shuyang Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
21
|
De Pasquale C, Campana S, Bonaccorsi I, Carrega P, Ferlazzo G. ILC in chronic inflammation, cancer and targeting with biologicals. Mol Aspects Med 2021; 80:100963. [PMID: 33726947 DOI: 10.1016/j.mam.2021.100963] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022]
Abstract
Since their discovery, Innate Lymphoid Cells (ILC) have emerged as important effector cells, serving multiple roles in maintaining tissue homeostasis and responding to tissue insults. As such, dysregulations of their function and distribution have been observed in a variety of immune-mediated diseases, suggesting a specific role for ILC in the pathophysiology of several disorders including chronic inflammation and cancer. Here, we provide an updated view on ILC biology dissecting their pathological or protective contribution in chronic inflammatory diseases such as multiple sclerosis, inflammatory bowel diseases, psoriasis, rheumatoid arthritis, asthma and COPD, atherosclerosis, also exploring ILC role in tumor surveillance and progression. Throughout the review, we will also highlight how the potential dual role of these cells for protective or pathogenic immunity in many inflammatory diseases makes them interesting targets for the development of novel therapeutic strategies, particularly promising.
Collapse
Affiliation(s)
- Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy.
| |
Collapse
|
22
|
Li Y, Wang F, Imani S, Tao L, Deng Y, Cai Y. Natural Killer Cells: Friend or Foe in Metabolic Diseases? Front Immunol 2021; 12:614429. [PMID: 33717101 PMCID: PMC7943437 DOI: 10.3389/fimmu.2021.614429] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The worldwide epidemic of metabolic diseases, especially obesity and other diseases caused by it, has shown a dramatic increase in incidence. A great deal of attention has been focused on the underlying mechanisms of these pathological processes and potential strategies to solve these problems. Chronic inflammation initiated by abdominal adipose tissues and immune cell activation in obesity is the major cause of the consequent development of complications. In addition to adipocytes, macrophages and monocytes, natural killer (NK) cells have been verified to be vital components involved in shaping the inflammatory microenvironment, thereby leading to various obesity-related metabolic diseases. Here, we provide an overview of the roles of NK cells and the interactions of these cells with other immune and nonimmune cells in the pathological processes of metabolic diseases. Finally, we also discuss potential therapeutic strategies targeting NK cells to treat metabolic diseases.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an, China.,Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yue Cai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
23
|
Nettersheim FS, De Vore L, Winkels H. Vaccination in Atherosclerosis. Cells 2020; 9:cells9122560. [PMID: 33266027 PMCID: PMC7760548 DOI: 10.3390/cells9122560] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is the major underlying pathology of cardiovascular diseases that together are the leading cause of death worldwide. The formation of atherosclerotic plaques is driven by chronic vascular inflammation. Although several risk factors have been identified and significant progress in disease prevention and treatment has been made, no therapeutic agents targeting inflammation are clinically available. Recent clinical trials established the potential of anti-inflammatory therapies as a treatment of atherosclerosis. However, adverse impacts on host defense have raised safety concerns about these therapies. Scientific evidence during the past 40 years implicated an adaptive immune response against plaque-associated autoantigens in atherogenesis. Preclinical data have underscored the protective potential of immunization against such targets precisely and without the impairment of host defense. In this review, we discuss the current vaccination strategies against atherosclerosis, supposed mechanisms of action, therapeutic potential, and the challenges that must be overcome in translating this idea into clinical practice.
Collapse
|
24
|
Kumrić M, Tičinović Kurir T, Borovac JA, Božić J. The Role of Natural Killer (NK) Cells in Acute Coronary Syndrome: A Comprehensive Review. Biomolecules 2020; 10:E1514. [PMID: 33167533 PMCID: PMC7694449 DOI: 10.3390/biom10111514] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
With poor outcomes and an immense financial burden, acute coronary syndrome (ACS) and its ischemic repercussions still present a major global health problem. Unfavorable outcomes seem to be mainly due to adverse cardiac remodeling. Since the inflammatory response takes an important role in remodeling secondary to myocardial infarction (MI), and as inflammation in this manner has not been completely elucidated, we attempted to give rise to a further understanding of ACS pathophysiology. Hence, in this review, we integrated current knowledge of complex communication networks between natural killer (NK) cells and immune and resident heart cells in the context of ACS. Based on available data, the role of NK cells seems to be important in the infarcted myocardium, where it affects heart remodeling. On the other hand, in atherosclerotic plaque, NK cells seem to be mere passers-by, except in the case of chronic infections by atherogenic pathogens. In that case, NK cells seem to support proinflammatory milieu. NK cell research is challenging due to ethical reasons, convergent evolution, and phenotypic diversity among individuals. Therefore, we argue that further research of NK cells in ACS is valuable, given their therapeutic potential in improving postischemic heart remodeling.
Collapse
Affiliation(s)
- Marko Kumrić
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
| | - Tina Tičinović Kurir
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
- Endocrinology Clinic, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Josip A. Borovac
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
- Institute of Emergency Medicine of Split-Dalmatia County (ZHM SDZ), Spinčićeva 1, 21000 Split, Croatia
| | - Joško Božić
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
| |
Collapse
|
25
|
Zernecke A, Winkels H, Cochain C, Williams JW, Wolf D, Soehnlein O, Robbins CS, Monaco C, Park I, McNamara CA, Binder CJ, Cybulsky MI, Scipione CA, Hedrick CC, Galkina EV, Kyaw T, Ghosheh Y, Dinh HQ, Ley K. Meta-Analysis of Leukocyte Diversity in Atherosclerotic Mouse Aortas. Circ Res 2020; 127:402-426. [PMID: 32673538 PMCID: PMC7371244 DOI: 10.1161/circresaha.120.316903] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The diverse leukocyte infiltrate in atherosclerotic mouse aortas was recently analyzed in 9 single-cell RNA sequencing and 2 mass cytometry studies. In a comprehensive meta-analysis, we confirm 4 known macrophage subsets-resident, inflammatory, interferon-inducible cell, and Trem2 (triggering receptor expressed on myeloid cells-2) foamy macrophages-and identify a new macrophage subset resembling cavity macrophages. We also find that monocytes, neutrophils, dendritic cells, natural killer cells, innate lymphoid cells-2, and CD (cluster of differentiation)-8 T cells form prominent and separate immune cell populations in atherosclerotic aortas. Many CD4 T cells express IL (interleukin)-17 and the chemokine receptor CXCR (C-X-C chemokine receptor)-6. A small number of regulatory T cells and T helper 1 cells is also identified. Immature and naive T cells are present in both healthy and atherosclerotic aortas. Our meta-analysis overcomes limitations of individual studies that, because of their experimental approach, over- or underrepresent certain cell populations. Mass cytometry studies demonstrate that cell surface phenotype provides valuable information beyond the cell transcriptomes. The present analysis helps resolve some long-standing controversies in the field. First, Trem2+ foamy macrophages are not proinflammatory but interferon-inducible cell and inflammatory macrophages are. Second, about half of all foam cells are smooth muscle cell-derived, retaining smooth muscle cell transcripts rather than transdifferentiating to macrophages. Third, Pf4, which had been considered specific for platelets and megakaryocytes, is also prominently expressed in the main population of resident vascular macrophages. Fourth, a new type of resident macrophage shares transcripts with cavity macrophages. Finally, the discovery of a prominent innate lymphoid cell-2 cluster links the single-cell RNA sequencing work to recent flow cytometry data suggesting a strong atheroprotective role of innate lymphoid cells-2. This resolves apparent discrepancies regarding the role of T helper 2 cells in atherosclerosis based on studies that predated the discovery of innate lymphoid cells-2 cells.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Holger Winkels
- Heart Center, University Hospital Cologne, Cologne, Germany
- Clinic III for Internal Medicine, Department of Cardiology, University of Cologne, Cologne, Germany
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Wüzburg, Germany
| | - Jesse W. Williams
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN USA
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN USA
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Klinikum LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Stockholm, Sweden
| | - Clint S. Robbins
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S1A1, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S1A1, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Toronto, ON M5G1L7, Canada
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Inhye Park
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, USA
- Division of Cardioascular Medicine, University of Virginia School of Medicine, Charlottesville, USA
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Myron I. Cybulsky
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Corey A. Scipione
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Elena V. Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA USA
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Huy Q. Dinh
- La Jolla Institute for Immunology, La Jolla, CA USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA USA
- Department of Bioengineering, University of California San Diego, CA, USA
| |
Collapse
|
26
|
Xu J, Yang Y. Potential genes and pathways along with immune cells infiltration in the progression of atherosclerosis identified via microarray gene expression dataset re-analysis. Vascular 2020; 28:643-654. [PMID: 32379583 DOI: 10.1177/1708538120922700] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory process characterized by the accumulation and formation of lipid-rich plaques within the layers of the arterial wall. Although numerous studies have reported the underlying pathogenesis, no data-based studies have been conducted to analyze the potential genes and immune cells infiltration in the different stages of atherosclerosis via bioinformatics analysis. METHODS In this study, we downloaded GSE100927 and GSE28829 from NCBI-GEO database. Gene ontology and pathway enrichment were performed via the DAVID database. The protein interaction network was constructed via STRING. Enriched hub genes were analyzed by the Cytoscape software. The evaluation of the infiltrating immune cells in the dataset samples was performed by the CIBERSORT algorithm. RESULTS We identified 114 common upregulated differentially expressed genes and 22 common downregulated differentially expressed genes. (adjust p value < 0.01 and log FC ≥ 1). A cluster of 10 genes including CYBA, SLC11A1, FCER1G, ITGAM, ITGB2, CD53, ITGAX, VAMP8, CLEC5A, and CD300A were found to be significant. Through the deconvolution algorithm CIBERSORT, we analyzed the significant alteration of immune cells infiltration in the progression of atherosclerosis with the threshold of the Wilcoxon test at p value <0.05. CONCLUSIONS These results may reveal the underlying correlations between genes and immune cells in atherosclerosis, which enable us to investigate the novel insights for the development of treatments and drugs.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Beijing, China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Beijing, China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Bekkering S, Saner C, Riksen NP, Netea MG, Sabin MA, Saffery R, Stienstra R, Burgner DP. Trained Immunity: Linking Obesity and Cardiovascular Disease across the Life-Course? Trends Endocrinol Metab 2020; 31:378-389. [PMID: 32305098 DOI: 10.1016/j.tem.2020.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/17/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023]
Abstract
Obesity, a chronic inflammatory disease, is the most prevalent modifiable risk factor for cardiovascular disease. The mechanisms underlying inflammation in obesity are incompletely understood. Recent developments have challenged the dogma of immunological memory occurring exclusively in the adaptive immune system and show that the innate immune system has potential to be reprogrammed. This innate immune memory (trained immunity) is characterized by epigenetic and metabolic reprogramming of myeloid cells following endogenous or exogenous stimulation, resulting in enhanced inflammation to subsequent stimuli. Trained immunity phenotypes have now been reported for other immune and non-immune cells. Here, we provide a novel perspective on the putative role of trained immunity in mediating the adverse cardiovascular effects of obesity and highlight potential translational pathways.
Collapse
Affiliation(s)
- Siroon Bekkering
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Christoph Saner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Matthew A Sabin
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Rinke Stienstra
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - David P Burgner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
28
|
Zhao TX, Newland SA, Mallat Z. 2019 ATVB Plenary Lecture: Interleukin-2 Therapy in Cardiovascular Disease: The Potential to Regulate Innate and Adaptive Immunity. Arterioscler Thromb Vasc Biol 2020; 40:853-864. [PMID: 32078364 DOI: 10.1161/atvbaha.119.312287] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Regulatory T cells and type-2 innate lymphoid cells represent 2 subsets of immune cells, which have been shown in preclinical models to be important in atherosclerosis and myocardial repair. Regulatory T cells play a crucial role in immune homeostasis and tolerance via their interactions with effector T cells, dendritic cells, and monocytes/macrophages. They also utilize and secrete inhibitory cytokines, including interleukin 10 and transforming growth factor β, to regulate or suppress pathogenic immune responses. Type-2 innate lymphoid cells have an important role in type-2 immune responses and tissue repair through secreting interleukins 5 and 13, as well as a variety of biological mediators and growth factors. Intriguingly, interleukin-2 has emerged as a common cytokine, which can be harnessed to upregulate both cell types, and also has important translational consequences as clinical trials are ongoing for its use in cardiovascular disease. Here, we briefly review the biology of these regulatory immune cell types, discuss the preclinical and clinical evidence for their functions in cardiovascular disease, examine the prospects for clinical translation and current ongoing trials, and finally, postulate how overlap in the mechanisms of upregulation may be leveraged in future treatments for patients.
Collapse
Affiliation(s)
- Tian X Zhao
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (T.X.Z., S.A.N., Z.M.)
| | - Stephen A Newland
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (T.X.Z., S.A.N., Z.M.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (T.X.Z., S.A.N., Z.M.)
- Paris-Descartes Université, Inserm U970, France (Z.M.)
| |
Collapse
|
29
|
Olson NC, Sitlani CM, Doyle MF, Huber SA, Landay AL, Tracy RP, Psaty BM, Delaney JA. Innate and adaptive immune cell subsets as risk factors for coronary heart disease in two population-based cohorts. Atherosclerosis 2020; 300:47-53. [PMID: 32209232 DOI: 10.1016/j.atherosclerosis.2020.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/29/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Cell-mediated immunity is implicated in atherosclerosis. We evaluated whether innate and adaptive immune cell subsets in peripheral blood are risk factors for coronary heart disease. METHODS A nested case-cohort study (n = 2155) was performed within the Multi-Ethnic Study of Atherosclerosis (MESA) and the Cardiovascular Health Study (CHS). Cases of incident myocardial infarction (MI) and incident angina (n = 880 total cases) were compared with a cohort random sample (n = 1275). Immune cell phenotypes (n = 34, including CD14+ monocytes, natural killer cells, γδ T cells, CD4+, CD8+ and CD19+ lymphocyte subsets) were measured from cryopreserved cells by flow cytometry. Cox proportional hazards models with adjustment for cardiovascular disease risk factors were used to evaluate associations of cell phenotypes with incident MI and a composite phenotype of incident MI or incident angina (MI-angina) over a median 9.3 years of follow-up. Th1, Th2, Th17, T regulatory (CD4+CD25+CD127-), naive (CD4+CD45RA+), memory (CD4+CD45RO+), and CD4+CD28- cells were specified as primary hypotheses. In secondary analyses, 27 additional cell phenotypes were investigated. RESULTS After correction for multiple testing, there were no statistically significant associations of CD4+ naive, memory, CD28-, or T helper cell subsets with MI or MI-angina in MESA, CHS, or combined-cohort meta analyses. Null associations were also observed for monocyte subsets, natural killer cells, γδ T cells, CD19+ B cell and differentiated CD4+ and CD8+ cell subsets. CONCLUSIONS The proportions of peripheral blood monocyte and lymphocyte subsets are not strongly related to the future occurrence of MI or angina in adults free of autoimmune disease.
Collapse
Affiliation(s)
- Nels C Olson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA.
| | - Colleen M Sitlani
- Department of Medicine, University of Washington, Seattle, WA, USA; Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Margaret F Doyle
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Sally A Huber
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA; Department of Biochemistry, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA
| | - Bruce M Psaty
- Department of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA; Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA.
| | - Joseph A Delaney
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA; College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
30
|
Passos LSA, Lupieri A, Becker-Greene D, Aikawa E. Innate and adaptive immunity in cardiovascular calcification. Atherosclerosis 2020; 306:59-67. [PMID: 32222287 DOI: 10.1016/j.atherosclerosis.2020.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 02/20/2020] [Indexed: 12/19/2022]
Abstract
Despite the focus placed on cardiovascular research, the prevalence of vascular and valvular calcification is increasing and remains a leading contributor of cardiovascular morbidity and mortality. Accumulating studies provide evidence that cardiovascular calcification is an inflammatory disease in which innate immune signaling becomes sustained and/or excessive, shaping a deleterious adaptive response. The triggering immune factors and subsequent inflammatory events surrounding cardiovascular calcification remain poorly understood, despite sustained significant research interest and support in the field. Most studies on cardiovascular calcification focus on innate cells, particularly macrophages' ability to release pro-osteogenic cytokines and calcification-prone extracellular vesicles and apoptotic bodies. Even though substantial evidence demonstrates that macrophages are key components in triggering cardiovascular calcification, the crosstalk between innate and adaptive immune cell components has not been adequately addressed. The only therapeutic options currently used are invasive procedures by surgery or transcatheter intervention. However, no approved drug has shown prophylactic or therapeutic effectiveness. Conventional diagnostic imaging is currently the best method for detecting, measuring, and assisting in the treatment of calcification. However, these common imaging modalities are unable to detect early subclinical stages of disease at the level of microcalcifications; therefore, the vast majority of patients are diagnosed when macrocalcifications are already established. In this review, we unravel the current knowledge of how innate and adaptive immunity regulate cardiovascular calcification; and put forward differences and similarities between vascular and valvular disease. Additionally, we highlight potential immunomodulatory drugs with the potential to target calcification and propose avenues in need of further translational inquiry.
Collapse
Affiliation(s)
- Livia S A Passos
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adrien Lupieri
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dakota Becker-Greene
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Pathology, Sechenov First Moscow State Medical University, Moscow, 119992, Russia.
| |
Collapse
|
31
|
Abstract
There is now overwhelming experimental and clinical evidence that atherosclerosis is a chronic inflammatory disease. Lessons from genome-wide association studies, advanced in vivo imaging techniques, transgenic lineage tracing mice, and clinical interventional studies have shown that both innate and adaptive immune mechanisms can accelerate or curb atherosclerosis. Here, we summarize and discuss the pathogenesis of atherosclerosis with a focus on adaptive immunity. We discuss some limitations of animal models and the need for models that are tailored to better translate to human atherosclerosis and ultimately progress in prevention and treatment.
Collapse
Affiliation(s)
- Dennis Wolf
- From the Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (D.W.).,Faculty of Medicine, University of Freiburg, Germany (D.W.)
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (K.L.).,Department of Bioengineering, University of California San Diego, La Jolla (K.L.)
| |
Collapse
|
32
|
Bonaccorsi I, Spinelli D, Cantoni C, Barillà C, Pipitò N, De Pasquale C, Oliveri D, Cavaliere R, Carrega P, Benedetto F, Ferlazzo G. Symptomatic Carotid Atherosclerotic Plaques Are Associated With Increased Infiltration of Natural Killer (NK) Cells and Higher Serum Levels of NK Activating Receptor Ligands. Front Immunol 2019; 10:1503. [PMID: 31354703 PMCID: PMC6639781 DOI: 10.3389/fimmu.2019.01503] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/17/2019] [Indexed: 12/27/2022] Open
Abstract
A wide array of immune cells, including lymphocytes, is known to be present and to play a pathogenetic role in atherosclerotic lesions. However, limited information is currently available regarding the presence of Natural Killer (NK) cell subsets within vessel plaque, and more in general, regarding their role in human atherosclerosis. We evaluated the distribution of NK cells in human carotid atherosclerotic plaques, dissecting asymptomatic and symptomatic patients (identified as affected by stroke, transient ischemic attack, or amaurosis fugax within 6 months) with the aim of shedding light on the putative contribution of NK cells to the pathogenic process that leads to plaque instability and subsequent clinical complications. We observed that carotid plaques were consistently infiltrated by NK cells and, among them, CD56brightperforinlow NK cells were abundantly present and displayed different markers of tissue residency (i.e., CD103 CD69 and CD49a). Interestingly, carotid atherosclerotic plaques of symptomatic patients showed a higher content of NK cells and an increased ratio between CD56brightperforinlow NK cells and their CD56dimperforinhigh counterpart. NK cells isolated from plaques of symptomatic patients were also stronger producers of IFN-γ. Analysis of the expression of NK activating receptor ligands (including MICA/B, ULBP-3, and B7-H6) in atherosclerotic carotid plaques revealed that they were abundantly expressed by a HLA-DR+CD11c+ myeloid cell population resident in the plaques. Remarkably, sera of symptomatic patients contained significant higher levels of soluble ligands for NK activating receptors. Our observations indicate that CD56bright NK cells accumulate within human atherosclerotic lesions and suggest a possible contribution of NK cells to the process determining plaque instability.
Collapse
Affiliation(s)
- Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department Human Pathology, University of Messina, Messina, Italy.,Research Center Cell Factory UniMe, University of Messina, Messina, Italy
| | - Domenico Spinelli
- Unit of Vascular Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,IRCCS, Istituto Giannina Gaslini, Genoa, Italy
| | - Chiara Barillà
- Unit of Vascular Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Narayana Pipitò
- Unit of Vascular Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department Human Pathology, University of Messina, Messina, Italy
| | - Daniela Oliveri
- Research Center Cell Factory UniMe, University of Messina, Messina, Italy.,Clinical Pathology Unit, University Hospital - A.O.U. Policlinico G. Martino, Messina, Italy
| | - Riccardo Cavaliere
- Laboratory of Immunology and Biotherapy, Department Human Pathology, University of Messina, Messina, Italy.,Research Center Cell Factory UniMe, University of Messina, Messina, Italy.,Clinical Pathology Unit, University Hospital - A.O.U. Policlinico G. Martino, Messina, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department Human Pathology, University of Messina, Messina, Italy.,Research Center Cell Factory UniMe, University of Messina, Messina, Italy
| | - Filippo Benedetto
- Unit of Vascular Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Department Human Pathology, University of Messina, Messina, Italy.,Research Center Cell Factory UniMe, University of Messina, Messina, Italy.,Clinical Pathology Unit, University Hospital - A.O.U. Policlinico G. Martino, Messina, Italy
| |
Collapse
|
33
|
Winkels H, Ley K. Natural Killer Cells at Ease: Atherosclerosis Is Not Affected by Genetic Depletion or Hyperactivation of Natural Killer Cells. Circ Res 2019; 122:6-7. [PMID: 29301835 DOI: 10.1161/circresaha.117.312289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Holger Winkels
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., K.L.); and Department of Bioengineering, University of California San Diego, La Jolla (K.L.)
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., K.L.); and Department of Bioengineering, University of California San Diego, La Jolla (K.L.).
| |
Collapse
|
34
|
Abstract
There is now overwhelming experimental and clinical evidence that arteriosclerosis is a chronic inflammatory disease. Lessons learned from genome-wide association studies, advanced in vivo imaging techniques, transgenic lineage tracing mice models and clinical interventional studies have shown that both innate and adaptive immune mechanisms can accelerate or curb arteriosclerosis. This article summarizes and discusses the pathogenesis of arteriosclerosis with a focus on the role of the adaptive immune system. Some limitations of animal models are discussed and the need for models that are tailored to better translate to human atherosclerosis and ultimately progress in prevention and treatment are emphasized.
Collapse
Affiliation(s)
- D Wolf
- Abteilung für Kardiologie und Angiologie I, Universitäts-Herzzentrum Freiburg, Freiburg, Deutschland
- Medizinische Fakultät, Universität Freiburg, Freiburg, Deutschland
| | - K Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, 92037, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Poli A, Michel T, Patil N, Zimmer J. Revisiting the Functional Impact of NK Cells. Trends Immunol 2018; 39:460-472. [PMID: 29496432 DOI: 10.1016/j.it.2018.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/08/2017] [Accepted: 01/23/2018] [Indexed: 01/28/2023]
Abstract
Immune responses are critical for the maintenance of homeostasis but can also upset the equilibrium, depending on the context and magnitude of the response. Natural killer (NK) cells are well known for their important roles in antiviral and antitumor immune responses, and they are currently used, mostly under optimized forms, as immunotherapeutic agents against cancer. Nevertheless, with accumulating examples of deleterious effects of NK cells, it is paramount to consider their negative contributions. Here, we critically review and comment on the literature surrounding undesirable aspects of NK cell activity, focusing on situations where they play a harmful rather than a protective role.
Collapse
Affiliation(s)
- Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; These authors contributed equally to this work and share first authorship
| | - Tatiana Michel
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; These authors contributed equally to this work and share first authorship
| | - Neha Patil
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
| |
Collapse
|