1
|
Wang Y, Zou Y, Jiang Q, Li W, Chai X, Zhao T, Liu S, Yuan Z, Yu C, Wang T. Ox-LDL-induced CD80 + macrophages expand pro-atherosclerotic NKT cells via CD1d in atherosclerotic mice and hyperlipidemic patients. Am J Physiol Cell Physiol 2024; 326:C1563-C1572. [PMID: 38586879 DOI: 10.1152/ajpcell.00043.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Atherosclerosis is an inflammatory disease of blood vessels involving the immune system. Natural killer T (NKT) cells, as crucial components of the innate and acquired immune systems, play critical roles in the development of atherosclerosis. However, the mechanism and clinical relevance of NKT cells in early atherosclerosis are largely unclear. The study investigated the mechanism influencing NKT cell function in apoE deficiency-induced early atherosclerosis. Our findings demonstrated that there were higher populations of NKT cells and interferon-gamma (IFN-γ)-producing NKT cells in the peripheral blood of patients with hyperlipidemia and in the aorta, blood, spleen, and bone marrow of early atherosclerotic mice compared with the control groups. Moreover, we discovered that the infiltration of CD80+ macrophages and CD1d expression on CD80+ macrophages in atherosclerotic mice climbed remarkably. CD1d expression increased in CD80+ macrophages stimulated by oxidized low-density lipoprotein (ox-LDL) ex vivo and in vitro. Ex vivo coculture of macrophages with NKT cells revealed that ox-LDL-induced CD80+ macrophages presented lipid antigen α-Galcer (alpha-galactosylceramide) to NKT cells via CD1d, enabling NKT cells to express more IFN-γ. Furthermore, a greater proportion of CD1d+ monocytes and CD1d+CD80+ monocytes were found in peripheral blood of hyperlipidemic patients compared with that of healthy donors. Positive correlations were found between CD1d+CD80+ monocytes and NKT cells or IFN-γ+ NKT cells in hyperlipidemic patients. Our findings illustrated that CD80+ macrophages stimulated NKT cells to secrete IFN-γ via CD1d-presenting α-Galcer, which may accelerate the progression of early atherosclerosis. Inhibiting lipid antigen presentation by CD80+ macrophages to NKT cells may be a promising immune target for the treatment of early atherosclerosis.NEW & NOTEWORTHY This work proposed the ox-LDL-CD80+ monocyte/macrophage-CD1d-NKT cell-IFN-γ axis in the progression of atherosclerosis. The proinflammatory IFN-γ+ NKT cells are closely related to CD1d+CD80+ monocytes in hyperlipidemic patients. Inhibiting CD80+ macrophages to present lipid antigens to NKT cells through CD1d blocking may be a new therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Yin Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| | - Yao Zou
- Department of Pharmacy, People's Hospital of Chongqing Liangjiang New District, Chongqing, People's Republic of China
| | - Qingsong Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, People's Republic of China
| | - Wenming Li
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xinyu Chai
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| | - Tingrui Zhao
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Sichuan, People's Republic of China
| | - Siyi Liu
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, People's Republic of China
- Chongqing Pharmacodynamic Evaluation Engineering Technology Research Center, Chongqing, People's Republic of China
| |
Collapse
|
2
|
Chakrabarti R, Duddu S, Tiwari A, Naidu KT, Sharma P, Chakravorty N, Shukla PC. Natural Killer T cells and the invariant subset promote atherosclerosis: A meta-analysis. Life Sci 2023; 321:121620. [PMID: 37011534 DOI: 10.1016/j.lfs.2023.121620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
AIMS Natural Killer T (NKT) cells are reported to be both pro- and anti-atherosclerotic. With this meta-analysis, we evaluated the NKT population and their subsets in regulating the atherosclerotic disease in mice. MAIN METHODS Eighteen pre-clinical (mice, n = 1276) and 6 clinical observational studies (humans, n = 116) met the eligibility criteria for inclusion. Random effects model was used and standard mean difference (SMD) was calculated for the cell counts and aortic lesion area. KEY FINDINGS Lesion area decreased in the absence of whole NKT cell population (-1.33[95%CI, -2.14, -0.52]), and in the absence of only iNKT subset (-0.66[95%CI, -1.69, 0.37]). However, lesion area increased after over-expression/activation of iNKTs (1.40[95%CI, 0.28, 2.52]). Atherogenic diet (AD) or high fat diet (HFD) increased the number of NKT cells (2.51[95%CI, 1.42, 3.61]), whereas the iNKT cell numbers and iNKT cell-specific gene expression decreased in mice (-2.04[95%CI, -3.34, -0.75]) and atherosclerotic patients (-1.81[95 % CI, -2.89, -0.74]). SIGNIFICANCE Here we show that, NKT and iNKT cells promote atherosclerosis. In general, NKT cell population increases with the progression of the plaque in mice and the numbers of iNKT cells reduce once the disease is established both in mice and humans.
Collapse
|
3
|
Miao T, Wang T, Feng T, Yuan D, Guo Q, Xiong F, Yang Y, Liu L, He Z, Huang B, Zhao J. Activated invariant natural killer T cells infiltrate aortic tissue as key participants in abdominal aortic aneurysm pathology. Immunology 2021; 164:792-802. [PMID: 34379797 DOI: 10.1111/imm.13401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023] Open
Abstract
Adaptive immunity and innate immunity have been implicated in the pathogenesis of abdominal aortic aneurysm (AAA), and damage and remodelling in the tunica media are a focus of the aneurysm development. Thus, identification of key immune cells or molecules that might be targets for the treatment of AAA is critical. We characterized the innate immune cells in human AAA tissue specimens by flow cytometry and found that apart from other lymphocytes, many invariant natural killer T (iNKT) cells marked as CD3 and Va24Ja18 had invaded the aortic tissues and were numerous, especially in the tunica media. These infiltrating iNKT cells have a high expression of CD69, indicating a highly active function. We were interested in whether iNKT cells could be the drivers of media damage in AAA. To answer this question, we used an AAA mouse model induced by angiotensin II (Ang II) infusion, which can reproduce the inflammatory response of AAA in mouse, which was confirmed by RNAseq. The results showed that the incidence of AAA was significantly higher after administration of α-galactosylceramide (α-GalCer), a synthetic glycolipid that activates iNKT cells via CD1d, compared with the Ang II-induced AAA alone (61·54% vs 31·82%) in mice. Histopathological and immunofluorescent staining results showed significantly more severe inflammatory infiltration and pathological lesions in the Ang II+α-GalCer treatment group. These results are highly suggestive that activated iNKT cells greatly contribute to AAA development and that the control of the activation state in iNKT cells may represent an important therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Tianyu Miao
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Tiehao Wang
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Ting Feng
- Laboratory of infection and immunity, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ding Yuan
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Guo
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Fei Xiong
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Yi Yang
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Lihua Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhangyu He
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Bin Huang
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| | - Jichun Zhao
- Vascular Surgery of West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Dounousi E, Duni A, Naka KK, Vartholomatos G, Zoccali C. The Innate Immune System and Cardiovascular Disease in ESKD: Monocytes and Natural Killer Cells. Curr Vasc Pharmacol 2021; 19:63-76. [PMID: 32600233 DOI: 10.2174/1570161118666200628024027] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Adverse innate immune responses have been implicated in several disease processes, including cardiovascular disease (CVD) and chronic kidney disease (CKD). The monocyte subsets natural killer (NK) cells and natural killer T (NKT) cells are involved in innate immunity. Monocytes subsets are key in atherogenesis and the inflammatory cascade occurring in heart failure. Upregulated activity and counts of proinflammatory CD16+ monocyte subsets are associated with clinical indices of atherosclerosis, heart failure syndromes and CKD. Advanced CKD is a complex state of persistent systemic inflammation characterized by elevated expression of proinflammatory and pro-atherogenic CD14++CD16+ monocytes, which are associated with cardiovascular events and death both in the general population and among patients with CKD. Diminished NK cells and NKT cells counts and aberrant activity are observed in both coronary artery disease and end-stage kidney disease. However, evidence of the roles of NK cells and NKT cells in atherogenesis in advanced CKD is circumstantial and remains to be clarified. This review describes the available evidence regarding the roles of specific immune cell subsets in the pathogenesis of CVD in patients with CKD. Future research is expected to further uncover the links between CKD associated innate immune system dysregulation and accelerated CVD and will ideally be translated into therapeutic targets.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Anila Duni
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- 2nd Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece
| | - Carmine Zoccali
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| |
Collapse
|
5
|
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: 4.0] [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
|
6
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall and the primary underlying cause of cardiovascular disease. Data from in vivo imaging, cell-lineage tracing and knockout studies in mice, as well as clinical interventional studies and advanced mRNA sequencing techniques, have drawn attention to the role of T cells as critical drivers and modifiers of the pathogenesis of atherosclerosis. CD4+ T cells are commonly found in atherosclerotic plaques. A large body of evidence indicates that T helper 1 (TH1) cells have pro-atherogenic roles and regulatory T (Treg) cells have anti-atherogenic roles. However, Treg cells can become pro-atherogenic. The roles in atherosclerosis of other TH cell subsets such as TH2, TH9, TH17, TH22, follicular helper T cells and CD28null T cells, as well as other T cell subsets including CD8+ T cells and γδ T cells, are less well understood. Moreover, some T cells seem to have both pro-atherogenic and anti-atherogenic functions. In this Review, we summarize the knowledge on T cell subsets, their functions in atherosclerosis and the process of T cell homing to atherosclerotic plaques. Much of our understanding of the roles of T cells in atherosclerosis is based on findings from experimental models. Translating these findings into human disease is challenging but much needed. T cells and their specific cytokines are attractive targets for developing new preventive and therapeutic approaches including potential T cell-related therapies for atherosclerosis.
Collapse
Affiliation(s)
- Ryosuke Saigusa
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Holger Winkels
- 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, La Jolla, CA, USA.
| |
Collapse
|
7
|
Haybar H, Maleki Behzad M, Shahrabi S, Ansari N, Saki N. Expression of Blood Cells Associated CD Markers and Cardiovascular Diseases: Clinical Applications in Prognosis. Lab Med 2020; 51:122-142. [PMID: 31340048 DOI: 10.1093/labmed/lmz049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a major cause of mortality worldwide. The results of various studies have shown that abnormality in the frequency and function of blood cells can be involved in CVD complications. In this review, we have focused on abnormalities in the expression of the CD (cluster of differentiation) markers of blood cells to assess the association of these abnormalities with CVD prognosis. METHODS We identified the relevant literature through a PubMed search (1990-2018) of English-language articles using the terms "Cardiovascular diseases", "CD markers", "leukocytes", "platelets", and "endothelial cells". RESULTS There is a variety of mechanisms for the effect of CD-marker expressions on CVDs prognosis, ranging from proinflammatory processes to dysfunctional effects in blood cells. CONCLUSION Considering the possible effects of CD-marker expression on CVDs prognosis, particularly prognosis of acute myocardial infarction and atherosclerosis, long-term studies in large cohorts are required to identify the prognostic value of CD markers and to target them with appropriate therapeutic agents.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masumeh Maleki Behzad
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Ansari
- Isfahan Bone Metabolic Disorders Research Center, Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Baumer Y, Gutierrez-Huerta CA, Saxena A, Dagur PK, Langerman SD, Tamura K, Ceasar JN, Andrews MR, Mitchell V, Collins BS, Yu Q, Teague HL, Playford MP, Bleck CKE, Mehta NN, McCoy JP, Powell-Wiley TM. Immune cell phenotyping in low blood volumes for assessment of cardiovascular disease risk, development, and progression: a pilot study. J Transl Med 2020; 18:29. [PMID: 31952533 PMCID: PMC6966880 DOI: 10.1186/s12967-020-02207-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/02/2020] [Indexed: 12/28/2022] Open
Abstract
Background Cardiovascular disease (CVD) is the leading cause of death in the world. Given the role of immune cells in atherosclerosis development and progression, effective methods for characterizing immune cell populations are needed, particularly among populations disproportionately at risk for CVD. Results By using a variety of antibodies combined in one staining protocol, we were able to identify granulocyte, lymphocyte, and monocyte sub-populations by CD-antigen expression from 500 µl of whole blood, enabling a more extensive comparison than what is possible with a complete blood count and differential (CBC). The flow cytometry panel was established and tested in a total of 29 healthy men and women. As a proof of principle, these 29 samples were split by their race/ethnicity: African-Americans (AA) (N = 14) and Caucasians (N = 15). We found in accordance with the literature that AA had fewer granulocytes and more lymphocytes when compared to Caucasians, though the proportion of total monocytes was similar in both groups. Several new differences between AA and Caucasians were noted that had not been previously described. For example, AA had a greater proportion of platelet adhesion on non-classical monocytes when compared to Caucasians, a cell-to-cell interaction described as crucially important in CVD. We also examined our flow panel in a clinical population of AA women with known CVD risk factors (N = 20). Several of the flow cytometry parameters that cannot be measured with the CBC displayed correlations with clinical CVD risk markers. For instance, Framingham Risk Score (FRS) calculated for each participant correlated with immune cell platelet aggregates (PA) (e.g. T cell PA β = 0.59, p = 0.03 or non-classical monocyte PA β = 0.54, p = 0.02) after adjustment for body mass index (BMI). Conclusion A flow cytometry panel identified differences in granulocytes, monocytes, and lymphocytes between AA and Caucasians which may contribute to increased CVD risk in AA. Moreover, this flow panel identifies immune cell sub-populations and platelet aggregates associated with CVD risk. This flow cytometry panel may serve as an effective method for phenotyping immune cell populations involved in the development and progression of CVD.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Cristhian A Gutierrez-Huerta
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Ankit Saxena
- Flow Cytometry Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven D Langerman
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Kosuke Tamura
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Joniqua N Ceasar
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Marcus R Andrews
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Valerie Mitchell
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Billy S Collins
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Quan Yu
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA
| | - Heather L Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher K E Bleck
- Electron Microscopy Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Philip McCoy
- Flow Cytometry Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Building 10-CRC, Room 5-5332, Bethesda, MD, 20892, USA. .,Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
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
|
10
|
VanderLaan PA, Reardon CA, Cabana VG, Wang CR, Getz GS. Invariant Natural Killer T-Cells and Total CD1d Restricted Cells Differentially Influence Lipid Metabolism and Atherosclerosis in Low Density Receptor Deficient Mice. Int J Mol Sci 2019; 20:ijms20184566. [PMID: 31540125 PMCID: PMC6770011 DOI: 10.3390/ijms20184566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 11/16/2022] Open
Abstract
Natural killer T (NKT) cells are a distinct subset of lymphocytes that bridge the innate and adaptive immune response and can be divided into type I invariant NKT cells (iNKT) and type II NKT cells. The objective of this study is to examine the effects of NKT cell on lipid metabolism and the initiation and progression of atherosclerosis in LDL receptor deficient (LDLR−/−) mice. Mice were fed an atherogenic diet for 4 or 8 weeks and plasma lipids, lipoproteins, and atherosclerosis were measured. The selective absence of iNKT cells in Jα18−/−LDLR−/− mice led to an increase in plasma cholesterol levels in female mice. Transgenic Vα14tg/LDLR−/− mice with elevated numbers of iNKT cells had increased late atherosclerosis of the innominate artery, though absence of either iNKT cells or all NKT cells and other CD1d expressing cells had varying effects on atherosclerotic lesion burden in the ascending aortic arch and aortic root. These studies not only highlight the potential modulatory role played by NKT cells in atherosclerosis and lipid metabolism, but also raise the possibility that divergent roles may be played by iNKT and CD1d restricted cells such as type II NKT cells or other CD1d expressing cells.
Collapse
Affiliation(s)
- Paul A VanderLaan
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University, 633 Clark St, Evanston, IL 60208, USA.
| | - Godfrey S Getz
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
11
|
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
|
12
|
Kritikou E, van Duijn J, Nahon JE, van der Heijden T, Bouwman M, Groeneveldt C, Schaftenaar FH, Kröner MJ, Kuiper J, van Puijvelde GH, Bot I. Disruption of a CD1d-mediated interaction between mast cells and NKT cells aggravates atherosclerosis. Atherosclerosis 2019; 280:132-139. [DOI: 10.1016/j.atherosclerosis.2018.11.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 10/10/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
|
13
|
Sharma A, Lawry SM, Klein BS, Wang X, Sherer NM, Zumwalde NA, Gumperz JE. LFA-1 Ligation by High-Density ICAM-1 Is Sufficient To Activate IFN-γ Release by Innate T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2018; 201:2452-2461. [PMID: 30171164 DOI: 10.4049/jimmunol.1800537] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/16/2018] [Indexed: 11/19/2022]
Abstract
By binding to its ligand ICAM-1, LFA-1 is known to mediate both adhesion and costimulatory signaling for T cell activation. The constitutively high LFA-1 cell surface expression of invariant NKT (iNKT) cells has been shown to be responsible for their distinctive tissue homing and residency within ICAM-rich endothelial vessels. However, the functional impact of LFA-1 on the activation of iNKT cells and other innate T lymphocyte subsets has remained largely unexplored. In particular, it is not clear whether LFA-1 contributes to innate-like pathways of T cell activation, such as IFN-γ secretion in response to IL-12. Using a recombinant ICAM-1-Fc fusion protein to stimulate human iNKT cells in the absence of APCs, we show that LFA-1 engagement enhances their IL-12-driven IFN-γ production. Surprisingly, exposure to high densities of ICAM-1 was also sufficient to activate iNKT cell cytokine secretion independently of IL-12 and associated JAK/STAT signaling. LFA-1 engagement induced elevated cytoplasmic Ca2+ and rapid ERK phosphorylation in iNKT cells, and the resulting IFN-γ secretion was dependent on both of these pathways. Analysis of freshly isolated human PBMC samples revealed that a fraction of lymphocytes that showed elevated LFA-1 cell surface expression produced IFN-γ in response to plate-bound ICAM-1-Fc. A majority of the responding cells were T cells, with the remainder NK cells. The responding T cells included iNKT cells, MAIT cells, and Vδ2+ γδ T cells. These results delineate a novel integrin-mediated pathway of IFN-γ secretion that is a shared feature of innate lymphocytes.
Collapse
Affiliation(s)
- Akshat Sharma
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Stephanie M Lawry
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Bruce S Klein
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Xiaohua Wang
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Nathan M Sherer
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| | - Nicholas A Zumwalde
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| |
Collapse
|
14
|
Abstract
The development of stress drives a host of biological responses that include the overproduction of a family of proteins named heat shock proteins (HSPs), because they were initially studied after heat exposure. HSPs are evolutionarily preserved proteins with a high degree of interspecies homology. HSPs are intracellular proteins that also have extracellular expression. The primary role of HSPs is to protect cell function by preventing irreversible protein damage and facilitating molecular traffic through intracellular pathways. However, in addition to their chaperone role, HSPs are immunodominant molecules that stimulate natural as well as disease-related immune reactivity. The latter may be a consequence of molecular mimicry, generating cross-reactivity between human HSPs and the HSPs of infectious agents. Autoimmune reactivity driven by HSPs could also be the result of enhancement of the immune response to peptides generated during cellular injury and of their role in the delivery of peptides to the major histocompatibility complex in antigen-presenting cells. In humans, HSPs have been found to participate in the pathogenesis of a large number of diseases. This review is focused on the role of HSPs in atherosclerosis and essential hypertension.
Collapse
Affiliation(s)
- B Rodríguez-Iturbe
- 1 Instituto Venezolano de Investigaciones Científicas (IVIC-Zulia), Nephrology Service Hospital Universitario, Universidad del Zulia , Maracaibo, Venezuela
| | - R J Johnson
- 2 Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| |
Collapse
|
15
|
Bagchi S, Genardi S, Wang CR. Linking CD1-Restricted T Cells With Autoimmunity and Dyslipidemia: Lipid Levels Matter. Front Immunol 2018; 9:1616. [PMID: 30061888 PMCID: PMC6055000 DOI: 10.3389/fimmu.2018.01616] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/29/2018] [Indexed: 11/13/2022] Open
Abstract
Dyslipidemia, or altered blood lipid content, is a risk factor for developing cardiovascular disease. Furthermore, several autoimmune diseases, including systemic lupus erythematosus, psoriasis, diabetes, and rheumatoid arthritis, are correlated highly with dyslipidemia. One common thread between both autoimmune diseases and altered lipid levels is the presence of inflammation, suggesting that the immune system might act as the link between these related pathologies. Deciphering the role of innate and adaptive immune responses in autoimmune diseases and, more recently, obesity-related inflammation, have been active areas of research. The broad picture suggests that antigen-presenting molecules, which present self-peptides to autoreactive T cells, can result in either aggravation or amelioration of inflammation. However, very little is known about the role of self-lipid reactive T cells in dyslipidemia-associated autoimmune events. Given that a range of autoimmune diseases are linked to aberrant lipid profiles and a majority of lipid-specific T cells are reactive to self-antigens, it is important to examine the role of these T cells in dyslipidemia-related autoimmune ailments and determine if dysregulation of these T cells can be drivers of autoimmune conditions. CD1 molecules present lipids to T cells and are divided into two groups based on sequence homology. To date, most of the information available on lipid-reactive T cells comes from the study of group 2 CD1d-restricted natural killer T (NKT) cells while T cells reactive to group 1 CD1 molecules remain understudied, despite their higher abundance in humans compared to NKT cells. This review evaluates the mechanisms by which CD1-reactive, self-lipid specific T cells contribute to dyslipidemia-associated autoimmune disease progression or amelioration by examining available literature on NKT cells and highlighting recent progress made on the study of group 1 CD1-restricted T cells.
Collapse
Affiliation(s)
| | | | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| |
Collapse
|
16
|
Getz GS, Reardon CA. T Cells in Atherosclerosis in Ldlr-/- and Apoe-/- Mice. JOURNAL OF IMMUNOLOGICAL SCIENCES 2018; 2:69-76. [PMID: 30854522 PMCID: PMC6404748 DOI: 10.29245/2578-3009/2018/3.1144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is the underlying basis for most cardiovascular diseases. It is a chronic inflammation affecting the arterial intima and is promoted by hypercholesterolemia. Cells of both the innate and adaptive immune systems contribute to this inflammation with macrophages and T cells being the most abundant immune cells in the atherosclerotic plaques. In this review, we discuss the studies that examined the role of T cells and T cell subsets in Apoe-/- and Ldlr-/- murine models of atherosclerosis. While there is a general consensus that Th1 cells are pro-atherogenic and regulatory T cells are atheroprotective, the role of other subsets is more ambiguous. In addition, the results in the two models of atherosclerosis do not always yield similar results. Additional studies in the two murine models using cell specific gene manipulations are needed.
Collapse
Affiliation(s)
- Godfrey S. Getz
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Catherine A. Reardon
- Ben May Institute for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
17
|
Subramanian S, Goodspeed L, Wang S, Ding Y, O'Brien KD, Getz GS, Chait A, Reardon CA. Deficiency of Invariant Natural Killer T Cells Does Not Protect Against Obesity but Exacerbates Atherosclerosis in Ldlr -/- Mice. Int J Mol Sci 2018; 19:ijms19020510. [PMID: 29419749 PMCID: PMC5855732 DOI: 10.3390/ijms19020510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 01/27/2018] [Accepted: 01/29/2018] [Indexed: 12/30/2022] Open
Abstract
Obesity is a chronic inflammatory state characterized by altered levels of adipose tissue immune cell populations. Natural killer T (NKT) cells are CD1d restricted lymphocyte subsets that recognize lipid antigens whose level decreases in obese adipose tissue. However, studies in mice with deficiency or increased levels of NKT cells have yielded contradictory results, so the exact role of these cells in obesity and adipose tissue inflammation is not yet established. We previously showed that Ldlr−/− mice with excess invariant NKT (iNKT) cells demonstrate significant weight gain, adiposity, metabolic abnormalities, and atherosclerosis. The current study evaluates the effects of NKT cell deficiency on obesity, associated metabolic changes, and atherosclerosis in Jα18−/−Ldlr−/− (lacking iNKT cells) and Cd1d−/−Ldlr−/− (lacking invariant and type II NKT cells) mice, and control mice were fed an obesogenic diet (high fat, sucrose, cholesterol) for 16 weeks. Contrary to expectations, Ja18−/−Ldlr−/− mice gained significantly more weight than Ldlr−/− or Cd1d−/−Ldlr−/− mice, developed hypertriglyceridemia, and had worsened adipose tissue inflammation. All the mice developed insulin resistance and hepatic triglyceride accumulation. Ja18−/−Ldlr−/− mice also had increased atherosclerotic lesion area. Our findings suggest that iNKT cells exacerbates the metabolic, inflammatory, and atherosclerotic features of diet-induced obesity. Further work is required to unravel the paradox of an apparently similar effect of iNKT cell surplus and depletion on obesity.
Collapse
Affiliation(s)
- Savitha Subramanian
- Diabetes Obesity Center for Excellence, Division of Metabolism, Endocrinology and Nutrition, University of Washington, 850 Republican Street Box 35805, Seattle, WA 98109, USA.
| | - Leela Goodspeed
- Diabetes Obesity Center for Excellence, Division of Metabolism, Endocrinology and Nutrition, University of Washington, 850 Republican Street Box 35805, Seattle, WA 98109, USA.
| | - Shari Wang
- Diabetes Obesity Center for Excellence, Division of Metabolism, Endocrinology and Nutrition, University of Washington, 850 Republican Street Box 35805, Seattle, WA 98109, USA.
| | - Yilei Ding
- Diabetes Obesity Center for Excellence, Division of Metabolism, Endocrinology and Nutrition, University of Washington, 850 Republican Street Box 35805, Seattle, WA 98109, USA.
| | - Kevin D O'Brien
- Division of Cardiology, University of Washington, Seattle, WA 98195, USA.
| | - Godfrey S Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.
| | - Alan Chait
- Diabetes Obesity Center for Excellence, Division of Metabolism, Endocrinology and Nutrition, University of Washington, 850 Republican Street Box 35805, Seattle, WA 98109, USA.
| | | |
Collapse
|
18
|
van Puijvelde GHM, Foks AC, van Bochove RE, Bot I, Habets KLL, de Jager SC, ter Borg MND, van Osch P, Boon L, Vos M, de Waard V, Kuiper J. CD1d deficiency inhibits the development of abdominal aortic aneurysms in LDL receptor deficient mice. PLoS One 2018; 13:e0190962. [PMID: 29346401 PMCID: PMC5773169 DOI: 10.1371/journal.pone.0190962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/22/2017] [Indexed: 11/19/2022] Open
Abstract
An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.
Collapse
Affiliation(s)
- Gijs H. M. van Puijvelde
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- * E-mail:
| | - Amanda C. Foks
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rosemarie E. van Bochove
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kim L. L. Habets
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Saskia C. de Jager
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mariëtte N. D. ter Borg
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Puck van Osch
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
19
|
Zhang N, Zhang M, Liu RT, Zhang P, Yang CL, Yue LT, Li H, Li YK, Duan RS. Statins reduce the expressions of Tim-3 on NK cells and NKT cells in atherosclerosis. Eur J Pharmacol 2017; 821:49-56. [PMID: 29288118 DOI: 10.1016/j.ejphar.2017.12.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022]
Abstract
3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitors (statins) have an immuno-regulatory effect in addition to lowing-lipids. Accumulated evidence showed that the expressions of T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) on natural killer (NK) cells increased in atherosclerotic patients and animal models. In this study, 14 patients treated with rosuvastatin and 12 patients with atorvastatin for more than 3 months were included and 20 patients without statins treatment as control. Both statins treatment reduced the expressions of Tim-3 on NK cells and their subtypes, natural killer T (NKT) cells and CD3+ T cells, and increased the proportions of NKT cells among peripheral blood mononuclear cells, accompanied by the decreased levels of total cholesterol, low density lipoprotein, and increased ratios of high density lipoprotein to cholesterol. These may contribute to the functions of statins in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Na Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Min Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Ru-Tao Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Peng Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Chun-Lin Yang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Long-Tao Yue
- Central laboratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Heng Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Yong-Kang Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China.
| |
Collapse
|
20
|
van Puijvelde GH, Kuiper J. NKT cells in cardiovascular diseases. Eur J Pharmacol 2017; 816:47-57. [DOI: 10.1016/j.ejphar.2017.03.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/10/2017] [Accepted: 03/23/2017] [Indexed: 12/17/2022]
|
21
|
Bagchi S, He Y, Zhang H, Cao L, Van Rhijn I, Moody DB, Gudjonsson JE, Wang CR. CD1b-autoreactive T cells contribute to hyperlipidemia-induced skin inflammation in mice. J Clin Invest 2017; 127:2339-2352. [PMID: 28463230 DOI: 10.1172/jci92217] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/03/2017] [Indexed: 01/09/2023] Open
Abstract
A large proportion of human T cells are autoreactive to group 1 CD1 proteins, which include CD1a, CD1b, and CD1c. However, the physiological role of the CD1 proteins remains poorly defined. Here, we have generated a double-transgenic mouse model that expresses human CD1b and CD1c molecules (hCD1Tg) as well as a CD1b-autoreactive TCR (HJ1Tg) in the ApoE-deficient background (hCD1Tg HJ1Tg Apoe-/- mice) to determine the role of CD1-autoreactive T cells in hyperlipidemia-associated inflammatory diseases. We found that hCD1Tg HJ1Tg Apoe-/- mice spontaneously developed psoriasiform skin inflammation characterized by T cell and neutrophil infiltration and a Th17-biased cytokine response. Anti-IL-17A treatment ameliorated skin inflammation in vivo. Additionally, phospholipids and cholesterol preferentially accumulated in diseased skin and these autoantigens directly activated CD1b-autoreactive HJ1 T cells. Furthermore, hyperlipidemic serum enhanced IL-6 secretion by CD1b+ DCs and increased IL-17A production by HJ1 T cells. In psoriatic patients, the frequency of CD1b-autoreactive T cells was increased compared with that in healthy controls. Thus, this study has demonstrated the pathogenic role of CD1b-autoreactive T cells under hyperlipidemic conditions in a mouse model of spontaneous skin inflammation. As a large proportion of psoriatic patients are dyslipidemic, this finding is of clinical significance and indicates that self-lipid-reactive T cells might serve as a possible link between hyperlipidemia and psoriasis.
Collapse
Affiliation(s)
- Sreya Bagchi
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ying He
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hong Zhang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Liang Cao
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ildiko Van Rhijn
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Infectious Diseases and Immunology, School of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - D Branch Moody
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
22
|
Echeverri Tirado LC, Yassin LM. B cells interactions in lipid immune responses: implications in atherosclerotic disease. Lipids Health Dis 2017; 16:30. [PMID: 28166809 PMCID: PMC5295187 DOI: 10.1186/s12944-016-0390-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/14/2016] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is considered as an inflammatory and chronic disorder with an important immunologic component, which underlies the majority of cardiovascular diseases; condition that belongs to a group of noncommunicable diseases that to date and despite of prevention and treatment approaches, they remain as the main cause of death worldwide, with 17.5 million of deaths every year. The impact of lipids in human health and disease is taking center stage in research, due to lipotoxicity explained by elevated concentration of circulating lipids, in addition to altered adipose tissue metabolism, and aberrant intracellular signaling. Immune response and metabolic regulation are highly integrated systems and the proper function of each one is dependent on the other. B lymphocytes express a variety of receptors that can recognize foreign, endogenous or modified self-antigens, among them oxidized low density lipoproteins, which are the main antigens in atherosclerosis. Mechanisms of B cells to recognize, remove and present lipids are not completely clear. However, it has been reported that B cell can recognize/remove lipids through a range of receptors, such as LDLR, CD1d, FcR and SR, which might have an atheroprotector or proatherogenic role during the course of atherosclerotic disease. Pertinent literature related to these receptors was examined to inform the present conclusions.
Collapse
Affiliation(s)
| | - Lina M Yassin
- Facultad de Medicina, Universidad CES, Calle 10 A Nro. 22-04, Medellín, Colombia.
| |
Collapse
|
23
|
|
24
|
Smith E, Croca S, Waddington KE, Sofat R, Griffin M, Nicolaides A, Isenberg DA, Torra IP, Rahman A, Jury EC. Cross-talk between iNKT cells and monocytes triggers an atheroprotective immune response in SLE patients with asymptomatic plaque. Sci Immunol 2016; 1:1/6/eaah4081. [PMID: 28783690 DOI: 10.1126/sciimmunol.aah4081] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/19/2016] [Indexed: 11/02/2022]
Abstract
Accelerated atherosclerosis is a complication of the autoimmune rheumatic disease systemic lupus erythematosus (SLE). We questioned the role played by invariant natural killer T (iNKT) cells in this process because they not only are defective in autoimmunity but also promote atherosclerosis in response to CD1d-mediated lipid antigen presentation. iNKT cells from SLE patients with asymptomatic plaque (SLE-P) had increased proliferation and interleukin-4 production compared with those from SLE patients with no plaque. The anti-inflammatory iNKT cell phenotype was associated with dyslipidemia and was driven by altered monocyte phospholipid expression and CD1d-mediated cross-talk between iNKT cells and monocytes but not B cells. Healthy iNKT cells differentiated in the presence of healthy monocytes and SLE-P serum polarized macrophages toward an anti-inflammatory M2 phenotype. Conversely, patients with clinical cardiovascular disease had unresponsive iNKT cells and increased proinflammatory monocytes. iNKT cell function could link immune responses, lipids, and cardiovascular disease in SLE patients and, together with serum lipid taxonomy, help predict preclinical atherosclerosis in SLE patients.
Collapse
Affiliation(s)
- Edward Smith
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K
| | - Sara Croca
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K
| | - Kirsty E Waddington
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K.,Centre for Clinical Pharmacology, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Reecha Sofat
- Centre for Clinical Pharmacology, Department of Medicine, University College London, London W1CE 6JF, U.K
| | | | - Andrew Nicolaides
- Vascular Noninvasive Diagnostic Centre, London, U.K.,Department of Vascular Surgery, Imperial College, London, U.K.,Department of Vascular Surgery, Nicosia Medical School, University of Nicosia, Nicosia, Cyprus
| | - David A Isenberg
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K
| | - Ines Pineda Torra
- Centre for Clinical Pharmacology, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Anisur Rahman
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, U.K.
| |
Collapse
|
25
|
Cheng F, Twardowski L, Reifenberg K, Winter K, Canisius A, Pross E, Fan J, Schmitt E, Shultz LD, Lackner KJ, Torzewski M. Combined B, T and NK Cell Deficiency Accelerates Atherosclerosis in BALB/c Mice. PLoS One 2016; 11:e0157311. [PMID: 27564380 PMCID: PMC5001715 DOI: 10.1371/journal.pone.0157311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/29/2016] [Indexed: 12/17/2022] Open
Abstract
This study focused on the unique properties of both the Ldlr knockout defect (closely mimicking the human situation) and the BALB/c (C) inbred mouse strain (Th-2 slanted immune response). We generated two immunodeficient strains with severe combined B- and T-cell immunodeficiency with or without a complete lack of natural killer cells to revisit the role of adaptive immune responses on atherogenesis. C-Ldlr-/-Rag1-/- mice, which show severe combined B- and T-cell immunodeficiency and C-Ldlr-/-Rag1-/-Il2rg-/- mice, which combine the T- and B-cell defect with a complete lack of natural killer cells and inactivation of multiple cytokine signalling pathways were fed an atherogenic Western type diet (WTD). Both B6-Ldlr-/- and C-Ldlr-/- immunocompetent mice were used as controls. Body weights and serum cholesterol levels of both immunodeficient strains were significantly increased compared to C-Ldlr-/- controls, except for cholesterol levels of C-Ldlr-/-Rag1-/- double mutants after 12 weeks on the WTD. Quantification of the aortic sinus plaque area revealed that both strains of immunodeficient mice developed significantly more atherosclerosis compared to C-Ldlr-/- controls after 24 weeks on the WTD. Increased atherosclerotic lesion development in C-Ldlr-/-Rag1-/-Il2rg-/- triple mutants was associated with significantly increased numbers of macrophages and significantly decreased numbers of smooth muscle cells compared to both C-Ldlr-/- wild type and C-Ldlr-/-Rag1-/- double mutants pointing to a plaque destabilizing effect of NK cell loss. Collectively, the present study reveals a previously unappreciated complexity with regard to the impact of lymphocytes on lipoprotein metabolism and the role of lymphocyte subsets in plaque composition.
Collapse
Affiliation(s)
- Fei Cheng
- Dr. Margarete Fischer-Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Laura Twardowski
- Department of Laboratory Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Kurt Reifenberg
- Center for Preclinical Research, German Cancer Research Center, Heidelberg, Germany
| | - Kerstin Winter
- Dr. Margarete Fischer-Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Antje Canisius
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eva Pross
- Dr. Margarete Fischer-Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Edgar Schmitt
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Torzewski
- Department of Laboratory Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
26
|
Felley LE, Sharma A, Theisen E, Romero-Masters JC, Sauer JD, Gumperz JE. Human Invariant NKT Cells Induce IL-1β Secretion by Peripheral Blood Monocytes via a P2X7-Independent Pathway. THE JOURNAL OF IMMUNOLOGY 2016; 197:2455-64. [PMID: 27534556 DOI: 10.4049/jimmunol.1600790] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/19/2016] [Indexed: 01/02/2023]
Abstract
The cytokine IL-1β plays a central role in inflammatory responses that are initiated by microbial challenges, as well as in those that are due to endogenous processes (often called sterile inflammation). IL-1β secretion that occurs independently of microbial stimulation is typically associated with the presence of endogenous alarmins, such as extracellular ATP (an indicator of cytopathic damage). In this study, we show that IL-2-activated human invariant NKT (iNKT) cells stimulate the secretion of IL-1β protein by human peripheral blood monocytes in a manner that requires neither the presence of microbial compounds nor signaling through the extracellular ATP receptor P2X7 Monocyte IL-1β production was specifically induced by iNKT cells, because similarly activated polyclonal autologous T cells did not have this effect. Secretion of IL-1β protein occurred rapidly (within 3-4 h) and required cell contact between the iNKT cells and monocytes. Similar to IL-1β production induced by TLR stimulation, the iNKT-induced pathway appeared to entail a two-step process involving NF-κB signaling and IL1B gene transcription, as well as assembly of the NLRP3 inflammasome and activation of caspase-1. However, in contrast to the classical inflammasome-mediated pathway of IL-1β production, activation of monocytes via P2X7 was dispensable for iNKT-induced IL-1β secretion, and potassium efflux was not required. Moreover, the iNKT-induced effect involved caspase-8 activity, yet it induced little monocyte death. These results suggest that IL-2-activated human iNKT cells induce monocytes to produce IL-1β through a distinctive pathway that does not require the presence of microbial danger signals or alarmins associated with cytopathic damage.
Collapse
Affiliation(s)
- Laura E Felley
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Akshat Sharma
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Erin Theisen
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - James C Romero-Masters
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and
| |
Collapse
|
27
|
Felley L, Gumperz JE. Are human iNKT cells keeping tabs on lipidome perturbations triggered by oxidative stress in the blood? Immunogenetics 2016; 68:611-22. [PMID: 27393663 DOI: 10.1007/s00251-016-0936-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/25/2016] [Indexed: 01/26/2023]
Abstract
The central paradigm of conventional MHC-restricted T cells is that they respond specifically to foreign peptides, while displaying tolerance to self-antigens. In contrast, it is now becoming clear that a number of innate-like T cell subsets-CD1-restricted T cells, Vγ9Vδ2 T cells, and MAIT cells-may operate by different rules: rather than focusing on the recognition of specific foreign antigens, these T cells all appear to respond to alterations to lipid-related pathways. By monitoring perturbations to the "lipidome," these T cells may be able to spring into action to deal with physiological situations that are of self as well as microbial origin. iNKT cells are a prime example of this type of lipidome-reactive T cell. As a result of their activation by self lyso-phospholipid species that are generated downstream of blood lipid oxidation, human iNKT cells in the vasculature may respond sensitively to a variety of oxidative stresses. Some of the cytokines produced by activated iNKT cells have angiogenic effects (e.g., GM-CSF, IL-8), whereas others (e.g., IFN-γ) are pro-inflammatory factors that can propagate vascular pathology by influencing the functions of macrophages and dendritic cells. Consistent with this, evidence is accumulating that iNKT cells contribute to atherosclerosis, which is one of the most common inflammatory pathologies, and one that is integrally related to characteristics of the lipidome.
Collapse
Affiliation(s)
- Laura Felley
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706, USA.
| |
Collapse
|
28
|
Abstract
Atherosclerosis is a chronic inflammatory disease that is initiated by the retention and accumulation of cholesterol-containing lipoproteins, particularly low-density lipoprotein, in the artery wall. In the arterial intima, lipoprotein components that are generated through oxidative, lipolytic, and proteolytic activities lead to the formation of several danger-associated molecular patterns, which can activate innate immune cells as well as vascular cells. Moreover, self- and non-self-antigens, such as apolipoprotein B-100 and heat shock proteins, can contribute to vascular inflammation by triggering the response of T and B cells locally. This process can influence the initiation, progression, and stability of plaques. Substantial clinical and experimental data support that the modulation of adaptive immune system may be used for treating and preventing atherosclerosis. This may lead to the development of more selective and less harmful interventions, while keeping host defense mechanisms against infections and tumors intact. Approaches such as vaccination might become a realistic option for cardiovascular disease, especially if they can elicit regulatory T and B cells and the secretion of atheroprotective antibodies. Nevertheless, difficulties in translating certain experimental data into new clinical therapies remain a challenge. In this review, we discuss important studies on the function of T- and B-cell immunity in atherosclerosis and their manipulation to develop novel therapeutic strategies against cardiovascular disease.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Göran K Hansson
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
29
|
Linking CD11b (+) Dendritic Cells and Natural Killer T Cells to Plaque Inflammation in Atherosclerosis. Mediators Inflamm 2016; 2016:6467375. [PMID: 27051078 PMCID: PMC4804096 DOI: 10.1155/2016/6467375] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/30/2015] [Accepted: 01/12/2016] [Indexed: 11/25/2022] Open
Abstract
Atherosclerosis remains the leading cause of death and disability in our Western society. To investigate whether the dynamics of leukocyte (sub)populations could be predictive for plaque inflammation during atherosclerosis, we analyzed innate and adaptive immune cell distributions in blood, plaques, and lymphoid tissue reservoirs in apolipoprotein E-deficient (ApoE−/−) mice and in blood and plaques from patients undergoing endarterectomy. Firstly, there was predominance of the CD11b+ conventional dendritic cell (cDC) subset in the plaque. Secondly, a strong inverse correlation was observed between CD11b+ cDC or natural killer T (NKT) cells in blood and markers of inflammation in the plaque (including CD3, T-bet, CCR5, and CCR7). This indicates that circulating CD11b+ cDC and NKT cells show great potential to reflect the inflammatory status in the atherosclerotic plaque. Our results suggest that distinct changes in inflammatory cell dynamics may carry biomarker potential reflecting atherosclerotic lesion progression. This not only is crucial for a better understanding of the immunopathogenesis but also bares therapeutic potential, since immune cell-based therapies are emerging as a promising novel strategy in the battle against atherosclerosis and its associated comorbidities. The cDC-NKT cell interaction in atherosclerosis serves as a good candidate for future investigations.
Collapse
|
30
|
Affiliation(s)
- Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
31
|
Rosales C, Tang D, Geng YJ. CD1d serves as a surface receptor for oxidized cholesterol induction of peroxisome proliferator-activated receptor-γ. Atherosclerosis 2015; 239:224-31. [PMID: 25618030 DOI: 10.1016/j.atherosclerosis.2015.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The cluster of differentiation-1d (CD1d) recognizes and presents the lipid antigens to NK-T lymphocytes. Atherosclerotic lesions contain atherogenic lipids, mainly cholesterol and its oxides. Peroxisome proliferator-activated receptor-γ (PPARγ) is also known to exist in atherosclerotic lesions, participating in regulation of lipid metabolism. The current study tested whether CD1d acts as a surface receptor that mediates induction and activation of PPARγ by oxysterols commonly found in atherosclerotic lesions. METHODS AND RESULTS CD1d overexpression in HEK 293 cells transfected with CD1d cDNA was confirmed by fluorescence, flow cytometry, Western blotting and mRNA expression. Tritiated ((3)H) 7-ketocholesterol (7K) was used for lipid binding assays. Radioactive assessment demonstrated an increased 7K-binding activity HEK 293 cells with CD1d overexpression. The 7K binding could be blocked by another oxysterol, 25-hydroxycholesterol, but not by native free cholesterol. Addition of CD1d:IgG dimer protein or an anti-CD1d antibody, but not control IgG, significantly diminished 7K binding to CD1d-expressing HEK 293 cells. CD1d deficiency markedly diminished the 7K-binding in macrophages and smooth muscle cells. Western blot and gel shift assays demonstrated that CD1d-mediated 7K binding induced expression and activation of PPARγ. The PPARγ agonist PGJ2 enhances the 7K stimulatory effect on PPARγ expression and activity but the antagonist GW9662 inhibits the 7K effect on the CD1d-expressing cells. CONCLUSIONS CD1d acts as a cell surface receptor that recognizes and binds oxysterols and initializes a pathway connecting oxysterol binding to PPARγ activation.
Collapse
Affiliation(s)
- Corina Rosales
- Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center at Houston, USA
| | - Daming Tang
- Texas Heart Institute, Houston, TX 77030, USA
| | - Yong-Jian Geng
- Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center at Houston, USA; Texas Heart Institute, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Li Y, To K, Kanellakis P, Hosseini H, Deswaerte V, Tipping P, Smyth MJ, Toh BH, Bobik A, Kyaw T. CD4+ natural killer T cells potently augment aortic root atherosclerosis by perforin- and granzyme B-dependent cytotoxicity. Circ Res 2014; 116:245-54. [PMID: 25398236 DOI: 10.1161/circresaha.116.304734] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE CD4(+) natural killer T (NKT) cells augment atherosclerosis in apolipoprotein E-deficient (ApoE)(-/-) mice but their mechanisms of action are unknown. OBJECTIVES We investigated the roles of bystander T, B, and NK cells; NKT cell-derived interferon-γ, interleukin (IL)-4, and IL-21 cytokines; and NKT cell-derived perforin and granzyme B cytotoxins in promoting CD4(+) NKT cell atherogenicity. METHODS AND RESULTS Transfer of CD4(+) NKT cells into T- and B-cell-deficient ApoE(-/-)Rag2(-/-) mice augmented aortic root atherosclerosis by ≈75% that was ≈30% of lesions in ApoE(-/-) mice; macrophage accumulation similarly increased. Transferred NKT cells were identified in the liver and atherosclerotic lesions of recipient mice. Transfer of CD4(+) NKT cells into T-, B-cell-deficient, and NK cell-deficient ApoE(-/-)Rag2(-/-)γC(-/-) mice also augmented atherosclerosis. These data indicate that CD4(+) NKT cells can exert proatherogenic effects independent of other lymphocytes. To investigate the role of NKT cell-derived interferon-γ, IL-4, and IL-21 cytokines and perforin and granzyme B cytotoxins, CD4(+) NKT cells from mice deficient in these molecules were transferred into NKT cell-deficient ApoE(-/-)Jα18(-/-) mice. CD4(+) NKT cells deficient in IL-4, interferon-γ, or IL-21 augmented atherosclerosis in ApoE(-/-)Jα18(-/-) mice by ≈95%, ≈80%, and ≈70%, respectively. Transfer of CD4(+) NKT cells deficient in perforin or granzyme B failed to augment atherosclerosis. Apoptotic cells, necrotic cores, and proinflammatory VCAM-1 (vascular cell adhesion molecule) and MCP-1 (monocyte chemotactic protein) were reduced in mice receiving perforin-deficient NKT cells. CD4(+) NKT cells are twice as potent as CD4(+) T cells in promoting atherosclerosis. CONCLUSIONS CD4(+) NKT cells potently promote atherosclerosis by perforin and granzyme B-dependent apoptosis that increases postapoptotic necrosis and inflammation.
Collapse
Affiliation(s)
- Yi Li
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Kelly To
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Peter Kanellakis
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Hamid Hosseini
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Virginie Deswaerte
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Peter Tipping
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Mark J Smyth
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Ban-Hock Toh
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Alexander Bobik
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Tin Kyaw
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.).
| |
Collapse
|
33
|
Tincati C, Basilissi M, Sinigaglia E, Merlini E, Carpani G, Monforte AD, Marchetti G. Invariant natural killer T (iNKT) cells in HAART-treated, HIV-positive patients with bone and cardiovascular impairment. PLoS One 2014; 9:e110287. [PMID: 25329381 PMCID: PMC4201495 DOI: 10.1371/journal.pone.0110287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/16/2014] [Indexed: 11/19/2022] Open
Abstract
Background Invariant Natural Killer T (iNKT) cells represent a determinant in the course of infections and diseases, however, their role in the pathogenesis of non-infectious co-morbidities in HIV-positive patients is unknown. Methods Flow cytometry was used to investigate iNKT cell frequency, phenotype and function in HIV-infected patients on HAART with bone and/or cardiovascular disorders and in HIV-positive controls free from co-morbidities. Results iNKT cells from subjects with bone and cardiovascular impairment expressed high levels of CD161 and predominantly secreted TNF. iNKT cells from individuals with bone disease alone did not show any distinctive phenotypical or functional characteristics. The functional capacity of iNKT cells in patients with cardiovascular disorder was impaired with no cytokine release upon stimulation. Conclusion iNKT cells may have a role in non-infectious co-morbidities in treated HIV disease, possibly through the exacerbation of inflammation. Further studies are needed to investigate iNKT cells in the pathogenesis of non-communicable disorders in HIV infection.
Collapse
Affiliation(s)
- Camilla Tincati
- Department of Health Sciences, Clinic of Infectious Diseases and Tropical Medicine, “San Paolo” Hospital, University of Milan, Milan, Italy
| | - Matteo Basilissi
- Department of Health Sciences, Clinic of Infectious Diseases and Tropical Medicine, “San Paolo” Hospital, University of Milan, Milan, Italy
| | | | - Esther Merlini
- Department of Health Sciences, Clinic of Infectious Diseases and Tropical Medicine, “San Paolo” Hospital, University of Milan, Milan, Italy
| | | | - Antonella d’Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases and Tropical Medicine, “San Paolo” Hospital, University of Milan, Milan, Italy
| | - Giulia Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases and Tropical Medicine, “San Paolo” Hospital, University of Milan, Milan, Italy
- * E-mail:
| |
Collapse
|
34
|
Winkels H, Weber C, Lutgens E, Gerdes N. Atherosclerosis: cell biology and lipoproteins focus on iNKT cells and CD40/CD40L in atherosclerosis and metabolic disorders. Curr Opin Lipidol 2014; 25:408-9. [PMID: 25186203 DOI: 10.1097/mol.0000000000000120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Holger Winkels
- aInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University (LMU), Munich, Germany bDepartment of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
35
|
Ait-Oufella H, Sage AP, Mallat Z, Tedgui A. Adaptive (T and B cells) immunity and control by dendritic cells in atherosclerosis. Circ Res 2014; 114:1640-60. [PMID: 24812352 DOI: 10.1161/circresaha.114.302761] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From INSERM UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (H.A.-O., Z.M., A.T.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France (H.A.-O.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | | | | | | |
Collapse
|
36
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
37
|
Legein B, Temmerman L, Biessen EAL, Lutgens E. Inflammation and immune system interactions in atherosclerosis. Cell Mol Life Sci 2013; 70:3847-69. [PMID: 23430000 PMCID: PMC11113412 DOI: 10.1007/s00018-013-1289-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 16.7 million deaths each year. The underlying cause of the majority of CVD is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. Today's picture is far more complex. Atherosclerosis is considered a chronic inflammatory disease that results in the formation of plaques in large and mid-sized arteries. Both cells of the innate and the adaptive immune system play a crucial role in its pathogenesis. By transforming immune cells into pro- and anti-inflammatory chemokine- and cytokine-producing units, and by guiding the interactions between the different immune cells, the immune system decisively influences the propensity of a given plaque to rupture and cause clinical symptoms like myocardial infarction and stroke. In this review, we give an overview on the newest insights in the role of different immune cells and subtypes in atherosclerosis.
Collapse
Affiliation(s)
- Bart Legein
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lieve Temmerman
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Erik A. L. Biessen
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, Pettenkoferstrasse 8a/9, 80336 Munich, Germany
| |
Collapse
|
38
|
Subramanian S, Turner MS, Ding Y, Goodspeed L, Wang S, Buckner JH, O'Brien K, Getz GS, Reardon CA, Chait A. Increased levels of invariant natural killer T lymphocytes worsen metabolic abnormalities and atherosclerosis in obese mice. J Lipid Res 2013; 54:2831-41. [PMID: 23922382 DOI: 10.1194/jlr.m041020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Obesity is a chronic inflammatory state characterized by infiltration of adipose tissue by immune cell populations, including T lymphocytes. Natural killer T (NKT) cells, a specialized lymphocyte subset recognizing lipid antigens, can be pro- or anti-inflammatory. Their role in adipose inflammation continues to be inconclusive and contradictory. In obesity, the infiltration of tissues by invariant NKT (iNKT) cells is decreased. We therefore hypothesized that an excess iNKT cell complement might improve metabolic abnormalities in obesity. Vα14 transgenic (Vα14tg) mice, with increased iNKT cell numbers, on a LDL receptor-deficient (Ldlr(-/-)) background and control Ldlr(-/-) mice were placed on an obesogenic diet for 16 weeks. Vα14tg.Ldlr(-/-) mice gained 25% more weight and had increased adiposity than littermate controls. Transgenic mice also developed greater dyslipidemia, hyperinsulinemia, insulin resistance, and hepatic triglyceride accumulation. Increased macrophage Mac2 immunostaining and proinflammatory macrophage gene expression suggested worsened adipose inflammation. Concurrently, these mice had increased atherosclerotic lesion area and aortic inflammation. Thus, increasing the complement of iNKT cells surprisingly exacerbated the metabolic, inflammatory, and atherosclerotic features of obesity. These findings suggest that the reduction of iNKT cells normally observed in obesity may represent a physiological attempt to compensate for this inflammatory condition.
Collapse
Affiliation(s)
- Savitha Subramanian
- Division of Metabolism, Endocrinology and Nutrition and University of Washington, Seattle, WA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bondarenko S, Catapano AL, Norata GD. The CD1d-natural killer T cell axis in atherosclerosis. J Innate Immun 2013; 6:3-12. [PMID: 23774666 DOI: 10.1159/000351034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 03/29/2013] [Indexed: 01/19/2023] Open
Abstract
A key role for 'lipid-sensing' CD1-restricted natural killer T (NKT) cells in the pathogenesis of atherosclerosis has been suggested. However, the biology of NKT cells remains poorly characterized, as in different experimental settings their activation was reported to both stimulate and suppress innate and adaptive immune responses. Most of the data from experimental models suggest that NKT cells are proatherogenic; however, it is debated whether the increase in atherosclerosis observed following NKT cell stimulation is a consequence of the inability to induce functional NKT cells rather than the proatherogenic nature of NKT cells. CD1d-expressing antigen-presenting cells and NKT cells were detected in mouse and human atherosclerotic lesions. Furthermore, several lysophospholipids and glycosphingolipids, known to accumulate in atherosclerotic plaques, are antigenic for human NKT cell clones. Lipid transfer proteins, such as apolipoprotein E and microsomal triglyceride transfer protein, are central to NKT cell responses. All these data suggest a profound relation between lipid metabolism, CD1d-NKT cell axis activation and atherosclerosis. In this review, we summarize the advances and gaps in our knowledge of NKT cell biology in the context of atherosclerosis as well as the possibility of influencing NKT cell polarization toward an atheroprotective phenotype.
Collapse
Affiliation(s)
- Sergey Bondarenko
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | | |
Collapse
|
40
|
Nakou E, Babageorgakas P, Bouchliou I, Tziakas DN, Miltiades P, Spanoudakis E, Margaritis D, Kotsianidis I, Stakos DA. Statin-induced immunomodulation alters peripheral invariant natural killer T-cell prevalence in hyperlipidemic patients. Cardiovasc Drugs Ther 2012; 26:293-9. [PMID: 22441892 DOI: 10.1007/s10557-012-6387-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE To assess the difference in the prevalence of invariant Natural Killer T (iNKT) lymphocytes between hyperlipidemic and control individuals and to evaluate changes in iNKT cell levels after 6 months lipid lowering therapy. METHODS A total of 77 hyperlipidemic individuals (54 ± 5 years) were assigned to simvastatin 40 mg or ezetimibe 10 mg daily for 6 months. Fifty individuals with normal cholesterol levels were used as control. iNKT cells were measured by flow cytometry in peripheral blood. RESULTS Patients with hypercholesterolemia had significantly lower iNKT cell levels (percentage on the lymphocyte population) compared to control group (0.16 ± 0.04% vs 0.39 ± 0.08%, p = 0.03). iNKT cells significantly increased after 6 months treatment with simvastatin (from 0.15 ± 0.04% to 0.28 ± 0.11%, p = 0.03) but not with ezetimibe (from 0.16 ± 0.05% to 0.17 ± 0.06%, p = 0.55). Simvastatin treatment did not alter the activation status of iNKT cells as measured by HLA-DR expression. Changes of iNKT cells were independent from changes in total (r(2) = 0.009, p = 0.76) or LDL cholesterol (r(2) = 0.008, p = 0.78) reached by simvastatin. CONCLUSIONS Hyperlipidemic patients have reduced numbers of iNKT in peripheral circulation compared to individuals with normal cholesterol levels. Their number is increasing after long term administration of simvastatin 40 mg but not after ezetimibe.
Collapse
Affiliation(s)
- Evangelia Nakou
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sag D, Wingender G, Nowyhed H, Wu R, Gebre AK, Parks JS, Kronenberg M, Hedrick CC. ATP-binding cassette transporter G1 intrinsically regulates invariant NKT cell development. THE JOURNAL OF IMMUNOLOGY 2012; 189:5129-38. [PMID: 23100511 DOI: 10.4049/jimmunol.1201570] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ATP-binding cassette transporter G1 (ABCG1) plays a role in the intracellular transport of cholesterol. Invariant NKT (iNKT) cells are a subpopulation of T lymphocytes that recognize glycolipid Ags. In this study, we demonstrate that ABCG1 regulates iNKT cell development and functions in a cell-intrinsic manner. Abcg1(-/-) mice displayed reduced frequencies of iNKT cells in thymus and periphery. Thymic iNKT cells deficient in ABCG1 had reduced membrane lipid raft content, and showed impaired proliferation and defective maturation during the early stages of development. Moreover, we found that Abcg1(-/-) mice possess a higher frequency of Vβ7(+) iNKT cells, suggesting alterations in iNKT cell thymic selection. Furthermore, in response to CD3ε/CD28 stimulation, Abcg1(-/-) thymic iNKT cells showed reduced production of IL-4 but increased production of IFN-γ. Our results demonstrate that changes in intracellular cholesterol homeostasis by ABCG1 profoundly impact iNKT cell development and function.
Collapse
Affiliation(s)
- Duygu Sag
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Andoh Y, Ogura H, Satoh M, Shimano K, Okuno H, Fujii S, Ishimori N, Eshima K, Tamauchi H, Otani T, Nakai Y, Van Kaer L, Tsutsui H, Onoé K, Iwabuchi K. Natural killer T cells are required for lipopolysaccharide-mediated enhancement of atherosclerosis in apolipoprotein E-deficient mice. Immunobiology 2012; 218:561-9. [PMID: 22954709 DOI: 10.1016/j.imbio.2012.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 07/11/2012] [Accepted: 07/14/2012] [Indexed: 12/12/2022]
Abstract
Lipopolysaccharide (LPS) has been shown to accelerate atherosclerosis and to increase the prevalence of IL-4-producing natural killer T (NKT) cells in various tissues. However, the role of NKT cells in the development of LPS-induced atherosclerotic lesions has not been fully tested in NKT cell-deficient mice. Here, we examined the lesion development in apolipoprotein E knockout (apoE-KO) mice and apoE-KO mice on an NKT cell-deficient, CD1d knockout (CD1d-KO) background (apoE-CD1d double knockout; DKO). LPS (0.5 μg/g body weight/wk) or phosphate-buffered saline (PBS) was intraperitoneally administered to apoE-KO and DKO mice (8-wk old) for 5 wk and atherosclerotic lesion areas were quantified thereafter. Consistent with prior reports, NKT cell-deficient DKO mice showed milder atherosclerotic lesions than apoE-KO mice. Notably, LPS administration significantly increased the lesion size in apoE-KO, but not in DKO mice, compared to PBS controls. Our findings suggest that LPS, and possibly LPS-producing bacteria, aggravate the development of atherosclerosis primarily through NKT cell activation and subsequent collaboration with NK cells.
Collapse
Affiliation(s)
- Yasuhiro Andoh
- Division of Immunobiology, Research Section of Pathophysiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Humoral and cellular immune responses in atherosclerosis: Spotlight on B- and T-cells. Vascul Pharmacol 2012; 56:193-203. [DOI: 10.1016/j.vph.2012.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/17/2012] [Accepted: 01/28/2012] [Indexed: 01/20/2023]
|
44
|
Getz GS, Vanderlaan PA, Reardon CA. Natural killer T cells in lipoprotein metabolism and atherosclerosis. Thromb Haemost 2011; 106:814-9. [PMID: 21946866 DOI: 10.1160/th11-05-0336] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 08/23/2011] [Indexed: 01/28/2023]
Abstract
Cells of both the innate and adaptive immune system participate in the development of atherosclerosis, a chronic inflammatory disorder of medium and large arteries. Natural killer T (NKT) cells express surface markers characteristic of natural killer cells and conventional T cells and bridge the innate and adaptive immune systems. The development and activation of NKT cells is dependent upon CD1d, a MHC-class I-type molecule that presents lipids, especially glycolipids to the T cell receptors on NKT cells. There are two classes of NKT cells; invariant NKT cells that express a semi-invariant T cell receptor and variant NKT cells. This review summarises studies in murine models in which the effect of the activation, overexpression or deletion of NKT cells or only invariant NKT cells on atherosclerosis has been examined.
Collapse
Affiliation(s)
- G S Getz
- University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
45
|
Saggini A, Anogeianaki A, Maccauro G, Teté S, Salini V, Caraffa A, Conti F, Fulcheri M, Galzio R, Shaik-Dasthagirisaheb Y. Cholesterol, Cytokines and Diseases. Int J Immunopathol Pharmacol 2011; 24:567-81. [DOI: 10.1177/039463201102400303] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A high level of cholesterol is associated with obesity, cardiovascular diseases and atherosclerosis. Immune response in atherosclerosis is mediated by chemokines which attract monocytes, leading to the innate immune response characterised by the production of cytokines. The immunoregulatory cytokines are an important bridge between innate and adductive immunity. TH1 cytokines are involved as effector T cells in inflammatory response, while TH2 cytokines can be anti-inflammatory such as IL-10 and IL-4. It is well known that statins enhance the production of TH2 cytokines whereas the secretion of TH1 cytokines is suppressed. For this purpose, we studied the significance of anti-inflammatory effect and suppression of inflammation by statins. In this paper we revisited the role of cholesterol and cytokines IL-18, IL-10, IL-12, TNF-α, interferon-γ, and chemokines in inflammatory diseases.
Collapse
Affiliation(s)
- A. Saggini
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - A. Anogeianaki
- Physiology Department, School of Medicine, Aristotle University of Thessaloniki, Greece
| | - G. Maccauro
- Department of Orthopaedics, Catholic University of Rome, Rome, Italy
| | - S. Teté
- School of Dentistry, University of Chieti, Italy
| | - V. Salini
- Orthopaedics Division, University of Chieti, Chieti, Italy
| | - A. Caraffa
- Orthopaedics Division, University of Perugia, Perugia, Italy
| | - F. Conti
- Department of Gyneacology, “Santo Spirito” Hospital, Pescara, Italy
| | - M. Fulcheri
- Department of Clinical Psychology, University of Chieti, Italy
| | - R. Galzio
- Department of Health Sciences, University of L'Aquila, Italy
| | | |
Collapse
|
46
|
Perrins CJ, Bobryshev YV. Current advances in understanding of immunopathology of atherosclerosis. Virchows Arch 2010; 458:117-23. [DOI: 10.1007/s00428-010-1006-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 10/05/2010] [Accepted: 10/20/2010] [Indexed: 01/13/2023]
|
47
|
Kyriakakis E, Cavallari M, Andert J, Philippova M, Koella C, Bochkov V, Erne P, Wilson SB, Mori L, Biedermann BC, Resink TJ, De Libero G. Invariant natural killer T cells: linking inflammation and neovascularization in human atherosclerosis. Eur J Immunol 2010; 40:3268-79. [PMID: 21061446 DOI: 10.1002/eji.201040619] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 06/28/2010] [Accepted: 08/17/2010] [Indexed: 12/16/2022]
Abstract
Atherosclerosis, a chronic inflammatory lipid storage disease of large arteries, is complicated by cardiovascular events usually precipitated by plaque rupture or erosion. Inflammation participates in lesion progression and plaque rupture. Identification of leukocyte populations involved in plaque destabilization is important for effective prevention of cardiovascular events. This study investigates CD1d-expressing cells and invariant NKT cells (iNKT) in human arterial tissue, their correlation with disease severity and symptoms, and potential mechanisms for their involvement in plaque formation and/or destabilization. CD1d-expressing cells were present in advanced plaques in patients who suffered from cardiovascular events in the past and were most abundant in plaques with ectopic neovascularization. Confocal microscopy detected iNKT cells in plaques, and plaque-derived iNKT cell lines promptly produced proinflammatory cytokines when stimulated by CD1d-expressing APC-presenting α-galactosylceramide lipid antigen. Furthermore, iNKT cells were diminished in the circulating blood of patients with symptomatic atherosclerosis. Activated iNKT cell-derived culture supernatants showed angiogenic activity in a human microvascular endothelial cell line HMEC-1-spheroid model of in vitro angiogenesis and strongly activated human microvascular endothelial cell line HMEC-1 migration. This functional activity was ascribed to IL-8 released by iNKT cells upon lipid recognition. These findings introduce iNKT cells as novel cellular candidates promoting plaque neovascularization and destabilization in human atherosclerosis.
Collapse
Affiliation(s)
- Emmanouil Kyriakakis
- Laboratory for Signal Transduction, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Braun NA, Covarrubias R, Major AS. Natural killer T cells and atherosclerosis: form and function meet pathogenesis. J Innate Immun 2010; 2:316-24. [PMID: 20375560 DOI: 10.1159/000296915] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Accepted: 01/20/2010] [Indexed: 01/17/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by dyslipidemia and accumulation of lipids in the arterial intima, with activation of both innate and adaptive immunity. Reciprocally, dyslipidemia associated with atherosclerosis can perturb normal immune function. Natural killer T (NKT) cells are a specialized group of immune cells that share characteristics with both conventional T cells and natural killer cells. However, unlike these cells, NKT cells recognize glycolipid antigens and produce both pro- and anti-inflammatory cytokines upon activation. Because of these unique characteristics, NKT cells have recently been ascribed a role in the regulation of immunity and inflammation, including cardiovascular disease. In addition, NKT cells represent a bridge between dyslipidemia and immune regulation. This review summarizes the current knowledge of NKT cells and discusses the interplay between dyslipidemia and the normal functions of NKT cells and how this might modulate inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Nicole A Braun
- Department of Molecular Pathology, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA
| | | | | |
Collapse
|
49
|
Chiba A, Cohen N, Brigl M, Brennan PJ, Besra GS, Brenner MB. Rapid and reliable generation of invariant natural killer T-cell lines in vitro. Immunology 2010; 128:324-33. [PMID: 20067532 DOI: 10.1111/j.1365-2567.2009.03130.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Several tools have proved useful in the study of invariant natural killer T (iNKT) cells, including CD1d-deficient mice, J alpha281-deficient mice, synthetic lipid antigens and antigen-loaded CD1d tetramers. However, the generation and examination of long-term primary murine iNKT cell lines in vitro has been challenging. Here, we show the rapid generation of iNKT cell lines from splenic iNKT cells of V alpha14 T-cell receptor (TCR) transgenic (Tg) mice. These purified iNKT cells were stimulated by bone marrow-derived dendritic cells (BMDCs) loaded with alpha-galactosylceramide (alphaGalCer) and cultured with interleukin (IL)-2 and IL-7. iNKT cells proliferated dramatically, and the cell number exhibited a 100-fold increase within 2 weeks and a 10(5)-fold increase in 8 weeks after repeated stimulation with alphaGalCer. The iNKT cell lines consisted of iNKT cells expressing V beta chains including V beta8.1/8.2, V beta14, V beta10, V beta6 and V beta7, and responded to stimulation with alphaGalCer presented both by BMDCs and by plate-bound CD1d. In addition, the iNKT cell lines produced interferon (IFN)-gamma when activated by lipopolysaccharide (LPS) or CpG oligodeoxynucleotide (ODN)-stimulated BMDCs. Further, we show that iNKT cell lines produced cytokines in response to microbial antigens. In summary, high-yield iNKT cell lines were generated very rapidly and robustly expanded, and these iNKT cells responded to both TCR and cytokine stimulation in vitro. Given the desire to study primary iNKT cells for many purposes, these iNKT cell lines should provide an important tool for the study of iNKT cell subsets, antigen and TCR specificity, activation, inactivation and effector functions.
Collapse
Affiliation(s)
- Asako Chiba
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
50
|
Packard RRS, Lichtman AH, Libby P. Innate and adaptive immunity in atherosclerosis. Semin Immunopathol 2009; 31:5-22. [PMID: 19449008 DOI: 10.1007/s00281-009-0153-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Accepted: 05/04/2009] [Indexed: 12/21/2022]
Abstract
Atherosclerosis, a chronic inflammatory disorder, involves both the innate and adaptive arms of the immune response that mediate the initiation, progression, and ultimate thrombotic complications of atherosclerosis. Most fatal thromboses, which may manifest as acute myocardial infarction or ischemic stroke, result from frank rupture or superficial erosion of the fibrous cap overlying the atheroma, processes that occur in inflammatorily active, rupture-prone plaques. Appreciation of the inflammatory character of atherosclerosis has led to the application of C-reactive protein as a biomarker of cardiovascular risk and the characterization of the anti-inflammatory and immunomodulatory actions of the statin class of drugs. An improved understanding of the pathobiology of atherosclerosis and further studies of its immune mechanisms provide avenues for the development of future strategies directed toward better risk stratification of patients as well as the identification of novel anti-inflammatory therapies. This review retraces leukocyte subsets involved in innate and adaptive immunity and their contributions to atherogenesis.
Collapse
Affiliation(s)
- René R S Packard
- Leducq Center for Cardiovascular Research, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 7, Boston, MA 02115, USA
| | | | | |
Collapse
|