1
|
Yang Y, Bai D, Jiang L, Chen Y, Wang M, Wang W, Wang H, He Q, Bu G, Long J, Yuan D. Stilbene glycosides alleviate atherosclerosis partly by promoting lipophagy of dendritic cells. Int Immunopharmacol 2024; 143:113223. [PMID: 39357204 DOI: 10.1016/j.intimp.2024.113223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease resulting from lipid metabolism disorders and immune imbalances. Dendritic cells (DCs) are key cells that regulate adaptive and adaptive immunity. When DCs engulf excessive amounts lipids, their function is altered, thereby, accelerating the inflammatory process of AS. Cellular lipophagy serves to reduce lipid accumulation and maintain cellular lipid metabolism balance. In this study, we investigated the effectiveness of 2,3,5,4'-tetrahydroxystilbene 2-O-β-D-glucoside (TSG) in intervening in the promotion of DCs lipid accumulation by ox-LDL, as well as its role in downregulating lipophagy. Our findings indicate that TSG reduces the maturity of DCs and promotes the differentiation of T cells towards Treg, thereby correcting the imbalanced Treg/Th17. These effects of TSG are closely associated with its inhibition of the PI3K-AKT-mTOR signaling pathway. After administering TSG to ApoE-/- mice that were fed a high-fat diet, there was a noticeable decrease in harmful blood lipids found in the serum. Additionally, the imbalanced Treg/Th17 levels in the spleen were restored, and the levels of pro-inflammatory factor IL-6 and IL-17A in the serum decreased, while the level of anti-inflammatory factor IL-10 increased. Furthermore, the arterial DCs showed a decrease in P62 content. Ultimately, these changes resulted in a reduction in plaque area. It is worth noting that the autophagy inhibitor chloroquine significantly altered the effects of TSG on ApoE-/- mice. In conclusion, this study reveals that TSG can alleviate AS. This is partly achieved through the activation of autophagy in DCs. By intervening in the lipophagy of DCs, it is possible to regulate the immune function of these cells, which in turn helps control the inflammation associated with AS. This presents a potential method for intervening in AS.
Collapse
Affiliation(s)
- Yunjun Yang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Dandan Bai
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Linhong Jiang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Yanran Chen
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Mengyuan Wang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Wenxin Wang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Haixia Wang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Qiongshan He
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China
| | - Guirong Bu
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China; Department of Pharmacy, Wuxi Huishan Traditional Chinese Medicine Hospital, Huijing Road 188, Wuxi 214100, Jiangsu, PR China
| | - Jun Long
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China.
| | - Dongping Yuan
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Xianlin Dadao 138, Nanjing 210023, Jiangsu, PR China.
| |
Collapse
|
2
|
Marriott E, Singanayagam A, El-Awaisi J. Inflammation as the nexus: exploring the link between acute myocardial infarction and chronic obstructive pulmonary disease. Front Cardiovasc Med 2024; 11:1362564. [PMID: 38450367 PMCID: PMC10915015 DOI: 10.3389/fcvm.2024.1362564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), particularly following acute exacerbations (AE-COPD), significantly heightens the risks and mortality associated with acute myocardial infarction (AMI). The intersection of COPD and AMI is characterised by a considerable overlap in inflammatory mechanisms, which play a crucial role in the development of both conditions. Although extensive research has been conducted on individual inflammatory pathways in AMI and COPD, the understanding of thrombo-inflammatory crosstalk in comorbid settings remains limited. The effectiveness of various inflammatory components in reducing AMI infarct size or slowing COPD progression has shown promise, yet their efficacy in the context of comorbidity with COPD and AMI is not established. This review focuses on the critical importance of both local and systemic inflammation, highlighting it as a key pathophysiological connection between AMI and COPD/AE-COPD.
Collapse
Affiliation(s)
- Eloise Marriott
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aran Singanayagam
- MRC Centre for Molecular Bacteriology & Infection, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Juma El-Awaisi
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Kott KA, Chan AS, Vernon ST, Hansen T, Kim T, de Dreu M, Gunasegaran B, Murphy AJ, Patrick E, Psaltis PJ, Grieve SM, Yang JY, Fazekas de St Groth B, McGuire HM, Figtree GA. Mass cytometry analysis reveals altered immune profiles in patients with coronary artery disease. Clin Transl Immunology 2023; 12:e1462. [PMID: 37927302 PMCID: PMC10621005 DOI: 10.1002/cti2.1462] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/09/2023] [Accepted: 08/09/2023] [Indexed: 11/07/2023] Open
Abstract
Objective The importance of inflammation in atherosclerosis is well accepted, but the role of the adaptive immune system is not yet fully understood. To further explore this, we assessed the circulating immune cell profile of patients with coronary artery disease (CAD) to identify discriminatory features by mass cytometry. Methods Mass cytometry was performed on patient samples from the BioHEART-CT study, gated to detect 82 distinct cell subsets. CT coronary angiograms were analysed to categorise patients as having CAD (CAD+) or having normal coronary arteries (CAD-). Results The discovery cohort included 117 patients (mean age 61 ± 12 years, 49% female); 79 patients (68%) were CAD+. Mass cytometry identified changes in 15 T-cell subsets, with higher numbers of proliferating, highly differentiated and cytotoxic cells and decreases in naïve T cells. Five T-regulatory subsets were related to an age and gender-independent increase in the odds of CAD incidence when expressing CCR2 (OR 1.12), CCR4 (OR 1.08), CD38 and CD45RO (OR 1.13), HLA-DR (OR 1.06) and Ki67 (OR 1.22). Markers of proliferation and differentiation were also increased within B cells, while plasmacytoid dendritic cells were decreased. This combination of changes was assessed using SVM models in discovery and validation cohorts (area under the curve = 0.74 for both), confirming the robust nature of the immune signature detected. Conclusion We identified differences within immune subpopulations of CAD+ patients which are indicative of a systemic immune response to coronary atherosclerosis. This immune signature needs further study via incorporation into risk scoring tools for the precision diagnosis of CAD.
Collapse
Affiliation(s)
- Katharine A Kott
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Adam S Chan
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Stephen T Vernon
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Thomas Hansen
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
| | - Taiyun Kim
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Macha de Dreu
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Bavani Gunasegaran
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | | | - Ellis Patrick
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | | | - Stuart M Grieve
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Department of RadiologyRoyal Prince Alfred HospitalSydneyNSWAustralia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Jean Y Yang
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Barbara Fazekas de St Groth
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Helen M McGuire
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Gemma A Figtree
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| |
Collapse
|
4
|
Reitsema RD, Hesselink BC, Abdulahad WH, van der Geest KSM, Brouwer E, Heeringa P, van Sleen Y. Aberrant phenotype of circulating antigen presenting cells in giant cell arteritis and polymyalgia rheumatica. Front Immunol 2023; 14:1201575. [PMID: 37600779 PMCID: PMC10433739 DOI: 10.3389/fimmu.2023.1201575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Giant Cell Arteritis (GCA) and Polymyalgia Rheumatica (PMR) are overlapping inflammatory diseases. Antigen-presenting cells (APCs), including monocytes and dendritic cells (DCs), are main contributors to the immunopathology of GCA and PMR. However, little is known about APC phenotypes in the peripheral blood at the time of GCA/PMR diagnosis. Methods APCs among peripheral blood mononuclear cells (PBMCs) of treatment-naive GCA and PMR patients were compared to those in age- and sex-matched healthy controls (HCs) using flow cytometry (n=15 in each group). We identified three monocyte subsets, and three DC subsets: plasmacytoid DCs (pDCs), CD141+ conventional DCs (cDC1) and CD1c+ conventional DCs (cDC2). Each of these subsets was analyzed for expression of pattern recognition receptors (TLR2, TLR4), immune checkpoints (CD86, PDL1, CD40) and activation markers (HLA-DR, CD11c). Results t-SNE plots revealed a differential clustering of APCs between GCA/PMR and HCs. Further analyses showed shifts in monocyte subsets and a lower proportion of the small population of cDC1 cells in GCA/PMR, whereas cDC2 proportions correlated negatively with CRP (r=-0.52). Classical monocytes of GCA/PMR patients show reduced expression of TLR2, HLA-DR, CD11c, which was in contrast to non-classical monocytes that showed higher marker expression. Additionally, single cell RNA sequencing in GCA patients identified a number of differentially expressed genes related to inflammation and metabolism in APCs. Conclusion Circulating non-classical monocytes display an activated phenotype in GCA/PMR patients at diagnosis, whereas classical monocytes show reduced expression of activation markers. Whether these findings reflect APC migration patterns or the effects of long-term inflammation remains to be investigated.
Collapse
Affiliation(s)
- Rosanne D. Reitsema
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Bernd-Cornèl Hesselink
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Wayel H. Abdulahad
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
5
|
Sauter M, Langer HF. Targeting Cell-Specific Molecular Mechanisms of Innate Immunity in Atherosclerosis. Front Physiol 2022; 13:802990. [PMID: 35432000 PMCID: PMC9010538 DOI: 10.3389/fphys.2022.802990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanisms of innate immunity contribute to inflammation, one of the major underlying causes of atherogenesis and progression of atherosclerotic vessel disease. How immune cells exactly contribute to atherosclerosis and interact with molecules of cholesterol homeostasis is still a matter of intense research. Recent evidence has proposed a potential role of previously underappreciated cell types in this chronic disease including platelets and dendritic cells (DCs). The pathophysiology of atherosclerosis is studied in models with dysfunctional lipid homeostasis and several druggable molecular targets are derived from these models. Specific therapeutic approaches focussing on these immune mechanisms, however, have not been successfully introduced into everyday clinical practice, yet. This review highlights molecular insights into immune processes related to atherosclerosis and potential future translational approaches targeting these molecular mechanisms.
Collapse
Affiliation(s)
- M. Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - H. F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- Department of Cardiology, University Heart Center Luebeck, University Hospital, Luebeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
- *Correspondence: H. F. Langer,
| |
Collapse
|
6
|
Kubiszewska I, Gackowska L, Obrycki Ł, Wierzbicka A, Helmin-Basa A, Kułaga Z, Wiese-Szadkowska M, Michałkiewicz J, Litwin M. Distribution and maturation state of peripheral blood dendritic cells in children with primary hypertension. Hypertens Res 2022; 45:401-413. [PMID: 34916664 DOI: 10.1038/s41440-021-00809-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/18/2021] [Accepted: 10/28/2021] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) play an important role in T cell alterations in primary hypertension (PH). We determined the numbers and maturation markers of peripheral blood total DCs (tDCs), myeloid cells (mDCs), and plasmacytoid cells (pDCs) and their association with hypertension-mediated organ damage (HMOD) markers and selected immune parameters in 30 adolescents with white coat hypertension (WCH), 25 adolescents with PH and a group of 35 age- and sex-matched children with normotension. Using multicolor flow cytometry, expression of maturation markers (CD86 and CD83) in tDCs (Lin1-/HLA-DR+), myeloid DCs (Lin1-/HLA-DR+/CD11c+) (mDCs), and plasmacytoid DCs (Lin1-/HLA-DR+/CD123+) (pDCs) and the distribution of peripheral Th17-bearing and T-reg cells were defined. In subjects with hypertension, carotid intima-media thickness (cIMT), left ventricular mass index (LVMI), and pulse wave velocity (PWV) were assessed. Compared with WCH and subjects with normotension, subjects with hypertension had reduced tDC and pDC numbers, an increased percentage of mDC subsets, an elevated mDC/pDC ratio, an increased population of mature mDC and pDC subsets bearing CD83 of high density, a decreased subset of CD86-bearing pDCs, and increased expression of DC activation markers (HLA-DR, CD86), as well as CD11c (mDCs) and CD123 (pDCs). PWV, LVMI, and cIMT values correlated negatively with tDCs and pDCs and positively with mDC numbers. Expression of DC maturation/activation markers (CD83, CD86, HLA-DR, CD11c, and CD123) correlated positively with PWV. Certain DC characteristics of WCH subjects resembled those of PH subjects (decreased tDC frequency and upregulation of activation marker expression). These changes correlated with HMOD. WCH subjects presented a DC phenotype that was intermediate between the normotensive and hypertensive phenotypes.
Collapse
Affiliation(s)
- Izabela Kubiszewska
- Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Lidia Gackowska
- Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Łukasz Obrycki
- Department of Nephrology, Kidney Transplantation and Arterial Hypertension, The Children's Memorial Health Institute, Warsaw, Poland
| | - Aldona Wierzbicka
- Department of Biochemistry and Experimental Medicine, The Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Helmin-Basa
- Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Zbigniew Kułaga
- Department of Public Health, The Children's Memorial Health Institute, Warsaw, Poland
| | - Małgorzta Wiese-Szadkowska
- Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Jacek Michałkiewicz
- Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland.,Department of Microbiology and Clinical Immunology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Mieczysław Litwin
- Department of Nephrology, Kidney Transplantation and Arterial Hypertension, The Children's Memorial Health Institute, Warsaw, Poland.
| |
Collapse
|
7
|
Peterson EA, Sun J, Wang J. Leukocyte-Mediated Cardiac Repair after Myocardial Infarction in Non-Regenerative vs. Regenerative Systems. J Cardiovasc Dev Dis 2022; 9:63. [PMID: 35200716 PMCID: PMC8877434 DOI: 10.3390/jcdd9020063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Innate and adaptive leukocytes rapidly mobilize to ischemic tissues after myocardial infarction in response to damage signals released from necrotic cells. Leukocytes play important roles in cardiac repair and regeneration such as inflammation initiation and resolution; the removal of dead cells and debris; the deposition of the extracellular matrix and granulation tissue; supporting angiogenesis and cardiomyocyte proliferation; and fibrotic scar generation and resolution. By organizing and comparing the present knowledge of leukocyte recruitment and function after cardiac injury in non-regenerative to regenerative systems, we propose that the leukocyte response to cardiac injury differs in non-regenerative adult mammals such as humans and mice in comparison to cardiac regenerative models such as neonatal mice and adult zebrafish. Specifically, extensive neutrophil, macrophage, and T-cell persistence contributes to a lengthy inflammatory period in non-regenerative systems for adverse cardiac remodeling and heart failure development, whereas their quick removal supports inflammation resolution in regenerative systems for new contractile tissue formation and coronary revascularization. Surprisingly, other leukocytes have not been examined in regenerative model systems. With this review, we aim to encourage the development of improved immune cell markers and tools in cardiac regenerative models for the identification of new immune targets in non-regenerative systems to develop new therapies.
Collapse
Affiliation(s)
| | | | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (E.A.P.); (J.S.)
| |
Collapse
|
8
|
Sun Z, Liu X, Liu Y, Zhao X, Zang X, Wang F. Immunosuppressive effects of dimethyl fumarate on dendritic cell maturation and migration: a potent protector for coronary heart disease. Immunopharmacol Immunotoxicol 2022; 44:178-185. [PMID: 35016591 DOI: 10.1080/08923973.2021.2025245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs), as a bridge between innate and adaptive immunity, play key roles in atherogenesis, particularly in plaque rupture, the underlying pathophysiologic cause of myocardial infarction. Targeting DC functions, including maturation and migration to atherosclerotic plaques, may be a novel therapeutic approach to atherosclerotic disease. Dimethyl fumarate (DMF), an agent consisting of a combination of fumaric acid esters, in current study were found to be able to suppress DC maturation by reducing the expression of costimulatory molecules and MHC class II and by blocking cytokine secretion. In addition, DMF efficiently inhibited the migration of activated DCs in vitro and in vivo by reducing the expression of chemokine receptor 7 (CCR7). Additionally, DMF efficiently inhibited the expression of the costimulatory molecule CD86, as well as the chemokine receptor CCR7 and the C-X-C motif chemokine receptor 4 (CXCR4), in healthy donor-derived purified DCs that had been stimulated by ST-segment elevation myocardial infarction (STEMI) patient serum. This study points to the potent therapeutic value of DMF for protecting against cardiovascular disease by suppressing DC functions.
Collapse
Affiliation(s)
- Zikai Sun
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China.,Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xiaoqiang Liu
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Yu Liu
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xin Zhao
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Zang
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
9
|
Lee YY, Kim SD, Park SC, Rhee MH. Panax ginseng: Inflammation, platelet aggregation, thrombus formation, and atherosclerosis crosstalk. J Ginseng Res 2022; 46:54-61. [PMID: 35058727 PMCID: PMC8753522 DOI: 10.1016/j.jgr.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Ginseng has been widely studied due to its various therapeutic properties on various diseases such as cardiovascular disease (CVD). Cardiovascular disease has been canonically known to be caused by high levels of low-density lipoproteins (LDL) in the bloodstream, in addition to the impaired vasodilatory effects of cholesterol. However, current research on CVD has revealed a cascade of mechanisms involving a series of events that contribute to the progression of CVD. Although this has been elucidated and summarized in previous studies the detailed correlation between platelet aggregation and innate immunity that plays an important role in CVD progression has not been thoroughly summarized. Furthermore, immune cell subtypes also contribute to the progression of plaque formation in the subendothelial layer. Thrombus formation and the coagulation cascade also have a vital role in the progression of atherosclerosis. Hence, in this mini review we aim to elucidate, summarize, and propose the potent therapeutic effect of ginseng on CVD, mainly on platelet aggregation, plaque formation, and thrombus formation.
Collapse
|
10
|
Dendritic Cells and Antiphospholipid Syndrome: An Updated Systematic Review. Life (Basel) 2021; 11:life11080801. [PMID: 34440545 PMCID: PMC8400181 DOI: 10.3390/life11080801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/23/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by autoreactive B and T cells against β2-glycoprotein I (B2GPI), with vascular thrombosis or obstetrical complications. Dendritic cells (DCs) are crucial in the generation of autoimmunity. Here, we conducted a comprehensive systematic review on the relationship between DC and APS. We performed a literature search of PubMed as of 26 March 2021. A total of 33 articles were extracted. DCs are pivotal in inducing inflammatory responses and orchestrating adaptive immunity. DCs contribute to the local inflammation regarding vascular thrombosis or obstetrical complications. Both B2GPI and antiphospholipid antibodies (aPL) can promote antigen presentation by DCs and the generation or maintenance of autoimmunity. In addition, plasmacytoid DC activation is enhanced by aPL, thereby augmenting the inflammatory response. In line with these findings, DC modulation appears promising as a future treatment for APS. In conclusion, our review indicated the crucial role of DCs in the pathogenesis of APS. Deeper understanding of the complex relationship would help in developing new treatment strategies.
Collapse
|
11
|
Mohmmad‐Rezaei M, Arefnezhad R, Ahmadi R, Abdollahpour‐Alitappeh M, Mirzaei Y, Arjmand M, Ferns GA, Bashash D, Bagheri N. An overview of the innate and adaptive immune system in atherosclerosis. IUBMB Life 2020. [DOI: 10.1002/iub.2425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mina Mohmmad‐Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | - Reza Arefnezhad
- Halal Research Center of IRI, FDA Tehran Iran
- Department of Anatomy, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | | | - Yousef Mirzaei
- Department of Biogeosciences, Scientific Research Center Soran University Soran Iraq
| | - Mohammad‐Hassan Arjmand
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
- Cancer Research Center Shahrekord University of Medical Sciences Shahrekord Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education Sussex United Kingdom
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| |
Collapse
|
12
|
Kott KA, Vernon ST, Hansen T, de Dreu M, Das SK, Powell J, Fazekas de St Groth B, Di Bartolo BA, McGuire HM, Figtree GA. Single-Cell Immune Profiling in Coronary Artery Disease: The Role of State-of-the-Art Immunophenotyping With Mass Cytometry in the Diagnosis of Atherosclerosis. J Am Heart Assoc 2020; 9:e017759. [PMID: 33251927 PMCID: PMC7955359 DOI: 10.1161/jaha.120.017759] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Coronary artery disease remains the leading cause of death globally and is a major burden to every health system in the world. There have been significant improvements in risk modification, treatments, and mortality; however, our ability to detect asymptomatic disease for early intervention remains limited. Recent discoveries regarding the inflammatory nature of atherosclerosis have prompted investigation into new methods of diagnosis and treatment of coronary artery disease. This article reviews some of the highlights of the important developments in cardioimmunology and summarizes the clinical evidence linking the immune system and atherosclerosis. It provides an overview of the major serological biomarkers that have been associated with atherosclerosis, noting the limitations of these markers attributable to low specificity, and then contrasts these serological markers with the circulating immune cell subtypes that have been found to be altered in coronary artery disease. This review then outlines the technique of mass cytometry and its ability to provide high-dimensional single-cell data and explores how this high-resolution quantification of specific immune cell subpopulations may assist in the diagnosis of early atherosclerosis in combination with other complimentary techniques such as single-cell RNA sequencing. We propose that this improved specificity has the potential to transform the detection of coronary artery disease in its early phases, facilitating targeted preventative approaches in the precision medicine era.
Collapse
Affiliation(s)
- Katharine A Kott
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Stephen T Vernon
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Thomas Hansen
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Macha de Dreu
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia
| | - Souvik K Das
- Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia
| | - Joseph Powell
- Garvan-Weizmann Centre for Cellular Genomics Garvan Institute Sydney Australia.,UNSW Cellular Genomics Futures Institute University of New South Wales Sydney Australia
| | - Barbara Fazekas de St Groth
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| | - Belinda A Di Bartolo
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia
| | - Helen M McGuire
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| | - Gemma A Figtree
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| |
Collapse
|
13
|
Sun L, Zhang W, Zhao Y, Wang F, Liu S, Liu L, Zhao L, Lu W, Li M, Xu Y. Dendritic Cells and T Cells, Partners in Atherogenesis and the Translating Road Ahead. Front Immunol 2020; 11:1456. [PMID: 32849502 PMCID: PMC7403484 DOI: 10.3389/fimmu.2020.01456] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a chronic process associated with arterial inflammation, the accumulation of lipids, plaque formation in vessel walls, and thrombosis with late mortal complications such as myocardial infarction and ischemic stroke. Immune and inflammatory responses have significant effects on every phase of atherosclerosis. Increasing evidence has shown that both innate and adaptive “arms” of the immune system play important roles in regulating the progression of atherosclerosis. Accumulating evidence suggests that a unique type of innate immune cell, termed dendritic cells (DCs), play an important role as central instigators, whereas adaptive immune cells, called T lymphocytes, are crucial as active executors of the DC immunity in atherogenesis. These two important immune cell types work in pairs to establish pro-atherogenic or atheroprotective immune responses in vascular tissues. Therefore, understanding the role of DCs and T cells in atherosclerosis is extremely important. Here, in this review, we will present a complete overview, based on existing knowledge of these two cell types in the atherosclerotic microenvironment, and discuss some of the novel means of targeting DCs and T cells as therapeutic tactics for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Yanfang Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Shan Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Lei Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Lin Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Wei Lu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Minghui Li
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| |
Collapse
|
14
|
Pistulli R, Andreas E, König S, Drobnik S, Kretzschmar D, Rohm I, Lichtenauer M, Heidecker B, Franz M, Mall G, Yilmaz A, Schulze PC. Characterization of dendritic cells in human and experimental myocarditis. ESC Heart Fail 2020; 7:2305-2317. [PMID: 32619089 PMCID: PMC7524053 DOI: 10.1002/ehf2.12767] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
Aims Dendritic cells (DCs) are central mediators of adaptive immunity, and there is growing evidence of their role in myocardial inflammatory disease. We hypothesized that plasmacytoid and myeloid DCs are involved in the mechanisms of myocarditis and analysed these two main subtypes in human myocarditis subjects, as well as in a murine model of experimental autoimmune myocarditis (EAM). Methods and results Circulating DCs were analysed by flow cytometry in patients with acute myocarditis, dilated cardiomyopathy, and controls. Myocardial biopsies were immunostained for the presence of DCs and compared with non‐diseased controls. In a mouse model of acute myocarditis induced through synthetic cardiac myosine peptide injection, effects of immunomodulation including DC inhibition through MCS‐18 versus placebo treatment were tested at the peak of inflammation (Day 21), as well as 1 week later (partial recovery). Circulatory pDCs and mDCs were significantly reduced in myocarditis patients compared with controls (P < 0.01 for both) and remained so even after 6 months of follow‐up. Human myocarditis biopsies showed accumulation of pDCs (two‐fold CD304+/three‐fold CD123+, all P < 0.05) compared with controls. Myocardial pDCs and mDCs accumulated in EAM (P for both <0.0001). MCS‐18 treatment reduced pDC levels (P = 0.009), reduced myocardial inflammation (myocarditis score reduction from 2.6 to 1.8, P = 0.026), and improved ejection fraction (P = 0.03) in EAM at Day 21 (peak of inflammation). This effect was not observed during the partial recovery of inflammation on Day 28. Conclusions Circulating DCs are reduced in human myocarditis and accumulate in the inflamed myocardium. MCS‐18 treatment reduces DCs in EAM, leading to amelioration of inflammation and left ventricular remodelling during the acute phase of myocarditis. Our data further elucidate the role of DCs and their specific subsets in acute inflammatory cardiomyopathies.
Collapse
Affiliation(s)
- Rudin Pistulli
- Department of Cardiology I - Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Albert-Schweitzer-Campus 1, Gebäude A1, Münster, 48149, Germany
| | - Elise Andreas
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Jena, Germany
| | | | - Stefanie Drobnik
- Institute of Forensic Medicine, University of Jena, Jena, Germany
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Jena, Germany
| | - Ilonka Rohm
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Jena, Germany
| | | | - Bettina Heidecker
- Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Marcus Franz
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Jena, Germany
| | - Gita Mall
- Institute of Forensic Medicine, University of Jena, Jena, Germany
| | - Atilla Yilmaz
- Internal Medicine Clinic II, Elisabeth Hospital, Schmalkalden, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Jena, Germany
| |
Collapse
|
15
|
Kumar D, Lee B, Puan KJ, Lee W, Luis BS, Yusof N, Andiappan AK, Del Rosario R, Poschmann J, Kumar P, DeLibero G, Singhal A, Prabhakar S, De Yun W, Poidinger M, Rötzschke O. Resistin expression in human monocytes is controlled by two linked promoter SNPs mediating NFKB p50/p50 binding and C-methylation. Sci Rep 2019; 9:15245. [PMID: 31645609 PMCID: PMC6811637 DOI: 10.1038/s41598-019-51592-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Resistin is a key cytokine associated with metabolic and inflammatory diseases. Especially in East Asian populations, the expression levels are strongly influenced by genetic polymorphisms. Mechanisms and functional implications of this genetic control are still unknown. By employing reporter assays, EMSA, inhibition studies, bisulphite sequencing, ChIP-Seq and gene-editing we show that the p50/p50 homodimer known to act as repressor for a number of pro-inflammatory genes plays a central role in the genetic regulation of resistin in monocytes along with promoter methylation. In the common RETN haplotype p50/p50 constitutively dampens the expression by binding to the promoter. In an Asian haplotype variant however this interaction is disrupted by the A allele of rs3219175. The SNP is in very close linkage to rs34861192, a CpG SNP, located 280 bp upstream which provides an allele-specific C-methylation site. rs34861192 is located in a 100 bp region found to be methylated in the common but not in the Asian haplotype, resulting in the latter having a higher basal expression, which also associates with elevated histone acetylation (H3K27ac). Genotype associations within cohort data of 200 East Asian individuals revealed significant associations between this haplotype and the plasma levels of factors such as TGF-b, S100B, sRAGE and IL-8 as well as with myeloid DC counts. Thus, the common RETN haplotype is tightly regulated by the epigenetic mechanism linked to p50/p50-binding. This control is lost in the Asian haplotype, which may have evolved to balance the antagonistic RETN effects on pathogen protection vs. metabolic and inflammatory disease induction.
Collapse
Affiliation(s)
- Dilip Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | - Bernett Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Wendy Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Boris San Luis
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Ricardo Del Rosario
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA, 02142, USA
| | - Jeremie Poschmann
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Centre de Recherche en Transplantation et Immunologie, Université de Nantes, Nantes, France
| | - Pavanish Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Gennaro DeLibero
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amit Singhal
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore
| | - Wang De Yun
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| |
Collapse
|
16
|
The P-selectin and PSGL-1 axis accelerates atherosclerosis via activation of dendritic cells by the TLR4 signaling pathway. Cell Death Dis 2019; 10:507. [PMID: 31263109 PMCID: PMC6602970 DOI: 10.1038/s41419-019-1736-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/22/2022]
Abstract
P-selectin and dendritic cells (DCs) are associated with atherosclerosis. However, their interactions in this setting are undefined. Herein, we investigated the role of P-selectin and its receptor P-selectin glycoprotein ligand (PSGL)-1 on atherosclerosis via activation of DCs. In the current study, a total of 34 patients with ST elevation myocardial infarction (STEMI) and 34 healthy control subjects were enrolled. Serum concentration of P-selectin was higher and the myeloid DC/plasmacytoid DC (mDC/pDC) ratio was lower in STEMI patients than in normal individuals. Interestingly, in STEMI patients, P-selectin was decreased and the mDC/pDC ratio was increased at 5–7 days after successful percutaneous coronary intervention, as compared with values on admission. Serum P-selectin was inversely correlated with the mDC/pDC ratio. Moreover, ApoE−/−P−/− and ApoE−/−PSGL-1−/− mice developed small atherosclerotic plaques after feeding of a western diet for 12 weeks and DC infiltration was significantly reduced. P-selectin stimulation markedly induced phenotypic maturation, enhanced secretion of inflammatory cytokines, communication with T cells, and the adhesion and migration of DCs. In vivo, DC maturation was significantly attenuated in P-selectin and PSGL1 knockout mice under hypercholesterolemic and inflammatory conditions. These effects were associated with the activation of myeloid differentiation primary response 88 (MYD88)-dependent and MyD88-independent Toll-like receptor 4 (TLR4) signaling pathways. Taken together, binding of P-selectin to PSGL-1 on DCs contributes to atherosclerosis progression via DC activation via the TLR4 signaling pathway.
Collapse
|
17
|
Negative impact of proteinuria on circulating myeloid dendritic cells. Clin Exp Nephrol 2019; 23:928-938. [PMID: 30879162 PMCID: PMC6555847 DOI: 10.1007/s10157-019-01724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/01/2019] [Indexed: 11/26/2022]
Abstract
Background A decrease in absolute numbers (abs.) of circulating dendritic cells (DCs) and recruitment into target organs has been reported, but whether the level of proteinuria associates with circulating DC abs. has not been clarified. Methods We conducted a cross-sectional study of 210 patients with kidney disease aged 21–96 years who were admitted to our hospital for kidney biopsy in 2007–2010. For accuracy, the level of proteinuria was thoroughly measured by 24-h urine collection from patients in their admitted condition. The abs. of total DCs (tDCs), myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) was measured by three-color fluorescence-activated cell sorting (FACS). Patients were divided into four groups based upon the quartile of each DC abs. and one-way ANOVA, and multivariable-adjusted regression analyses were performed. Results Quantile analysis showed that the level of daily proteinuria decreased with increasing blood mDC abs., with mean proteinuria levels (g/day) of 2.45, 1.68, 1.68, 1.10 for those in mDC abs. quartiles ≤ 445, < 686, < 907, ≥ 907 cells/102 µL (p = 0.0277), respectively. Multivariate-adjusted regression analysis revealed that the mDC abs. was negatively associated with proteinuria (95% CI − 57.0 to − 8.5) and positively associated with male gender (95% CI 66.2–250.5). Independent associations were also shown between pDCs abs. and estimated glomerular filtration rate (eGFR) (95% CI 0.14–2.67) and C-reactive protein (95% CI − 49.4 to − 9.9) and between tDCs abs. and male gender (95% CI 54.5–253.6) and C-reactive protein (95% CI − 80.5 to − 13.4). Conclusion We first reported that circulating mDC abs. has a negative association with the level of proteinuria.
Collapse
|
18
|
Yetkin GI, Yucel AA, Tekin İÖ, Yılmaz M, Atalay H, Yetkin E. Dendritic cell activation is blunted in patients with coronary artery disease and diabetes mellitus. J Diabetes Complications 2019; 33:134-139. [PMID: 30522792 DOI: 10.1016/j.jdiacomp.2018.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 11/10/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND It has been shown that functional status of dendritic cells (DCs) in diabetic patients with unstable angina pectoris (UAP) are more mature and activated than diabetic patients without coronary artery disease (CAD) and none diabetic patients with UAP. Accordingly we aimed to assess the activation of DCs in patients with CAD with/and without Diabetes Mellitus (DM) and compare to those in subjects with normal coronary arteries (NCA). MATERIALS AND METHODS Twenty three patients with severe CAD who were scheduled to coronary artery by-pass grafting surgery and 6 patients with angiographycally NCAs were included in the study. Activation of peripheral blood DCs have been analyzed by flow cytometric measures of CD86 activation. RESULTS In patients with CAD and without DM, DC activation significantly increased after stimulation of oxidesized LDL (135 ± 121 vs 248 ± 197 p = 0.024). However this activation didn't significantly increased in patients with CAD and DM (100 ± 20 vs 120 ± 97, p = 0,54). Patients with NCAs and without DM showed marked activation of CD86 after stimulation with ox-LDL. CONCLUSION We have documented that DC activation, upon stimulation of ox-LDL has blunted in patients with CAD compared to patients with NCAs. Moreover this defective activation is more pronounced in those with diabetic patients with CAD.
Collapse
Affiliation(s)
| | - Aysegul Atak Yucel
- Gazi University Faculty of Medicine, Department of Immunology, Ankara, Turkey.
| | - İshak Özel Tekin
- Zonguldak Bulent Ecevit University School of Medicine, Department of Immonology, Zonguldak, Turkey
| | - Mustafa Yılmaz
- Cukurova University Faculty of Medicine, Department of Pediatric Allergy and Immunology, Adana, Turkey.
| | - Hakan Atalay
- Middle East Hospital Division of Cardiovascular Surgery, Mersin, Turkey.
| | - Ertan Yetkin
- Yenisehir Hospital Division of Cardiology, Mersin, Turkey.
| |
Collapse
|
19
|
Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol 2018; 114:4. [PMID: 30523422 PMCID: PMC6290728 DOI: 10.1007/s00395-018-0712-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Myocardial infarction triggers infiltration of several types of immune cells that coordinate both innate and adaptive immune responses. These play a dual role in post-infarction cardiac remodeling by initiating and resolving inflammatory processes, which needs to occur in a timely and well-orchestrated way to ensure a reestablishment of normalized cardiac functions. Thus, therapeutic modulation of immune responses might have benefits for infarct patients. While such strategies have shown great potential in treating cancer, applications in the post-infarction context have been disappointing. One challenge has been the complexity and plasticity of immune cells and their functions in cardiac regulation and healing. The types appear in patterns that are temporally and spatially distinct, while influencing each other and the surrounding tissue. A comprehensive understanding of the immune cell repertoire and their regulatory functions following infarction is sorely needed. Processes of cardiac remodeling trigger additional genetic changes that may also play critical roles in the aftermath of cardiovascular disease. Some of these changes involve non-coding RNAs that play crucial roles in the regulation of immune cells and may, therefore, be of therapeutic interest. This review summarizes what is currently known about the functions of immune cells and non-coding RNAs during post-infarction wound healing. We address some of the challenges that remain and describe novel therapeutic approaches under development that are based on regulating immune responses through non-coding RNAs in the aftermath of the disease.
Collapse
Affiliation(s)
- Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Dodsworth
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
20
|
Manthey H, Zernecke A. Dendritic cells in atherosclerosis: Functions in immune regulation and beyond. Thromb Haemost 2017; 106:772-8. [DOI: 10.1160/th11-05-0296] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 08/02/2011] [Indexed: 12/15/2022]
Abstract
SummaryChronic inflammation drives the development of atherosclerosis. Dendritic cells (DCs) are known as central mediators of adaptive immune responses and the development of immunological memory and tolerance. DCs are present in non-diseased arteries, and accumulate within atherosclerotic lesions where they can be localised in close vicinity to T cells. Recent work has revealed important functions of DCs in regulating immune mechanisms in atherogenesis, and vaccination strategies using DCs have been explored for treatment of disease. However, in line with a phenotypical and functional overlap with plaque macrophages vascular DCs were also identified to engulf lipids, thus contributing to lipid burden in the vessel wall and initiation of lesion growth. Furthermore, a function of DCs in regulating cholesterol homeostasis has been revealed. Finally, phenotypically distinct plasmacytoid dendritic cells (pDCs) have been identified within atherosclerotic lesions. This review will dissect the multifaceted contribution of DCs and pDCs to the initiation and progression of atherosclerosis and the experimental approaches utilising DCs in therapeutic vaccination strategies.
Collapse
|
21
|
Comparison of circulating dendritic cell and monocyte subsets at different stages of atherosclerosis: insights from optical coherence tomography. BMC Cardiovasc Disord 2017; 17:270. [PMID: 29047360 PMCID: PMC5648428 DOI: 10.1186/s12872-017-0702-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND While specific patterns of circulating dendritic cells (DCs) and monocytes are associated with the incidence of coronary artery disease, the characterization of circulating DC and monocyte subsets in patients with different stages of atherosclerosis remains unclear. METHODS Forty-eight patients with unstable angina pectoris (UAP) diagnosed by angiography were enrolled. Likewise, 31 patients with ST-segment elevation myocardial infarction (STEMI) were enrolled and confirmed with the presence of thrombosis by angiography. Plaque features of 48 UAP patients were evaluated at the culprit lesions by OCT. Circulating myeloid DCs (mDCs), plasmacytoid DCs (pDCs) and monocyte subsets were analyzed using flow cytometry. RESULTS The proportions and absolute counts of mDC2s, which specifically express CD141 and possess the ability to activate CD8+ T lymphocytes, significantly decreased in patients with UAP and STEMI when compared with controls (0.08 × 104 ± 0.05 × 104/ml and 0.08 × 104 ± 0.06 × 104/ml vs. 0.11 × 104 ± 0.06 × 104/ml, p = 0.027). On the other hand, patients with UAP and STEMI had significantly higher proportions and counts of Mon2 subsets. In the OCT subgroup, patients with thin-cap fibroatheroma (TCFA) had higher proportions and absolute number of Mon2 (11.96% ± 4.27% vs. 9.42% ± 4.05%, p = 0.034; 5.17 × 104/ml ± 1.92 × 104/ml vs. 3.53 × 104/ml ± 2.65 × 104/ml, p = 0.045) than those without TCFA. However, there was no remarkable difference in mDC2s between patients with and without TCFA. CONCLUSIONS Circulating Mon2 appears to be a promising marker for the severity of atherosclerotic plaque.
Collapse
|
22
|
Kim JU, Kim M, Kim S, Nguyen TT, Kim E, Lee S, Kim S, Kim H. Dendritic Cell Dysfunction in Patients with End-stage Renal Disease. Immune Netw 2017; 17:152-162. [PMID: 28680376 PMCID: PMC5484645 DOI: 10.4110/in.2017.17.3.152] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
End-stage renal disease (ESRD) with immune disorder involves complex interactions between the innate and adaptive immune responses. ESRD is associated with various alterations in immune function such as a reduction in polymorphonuclear leukocyte bactericidal activity, a suppression of lymphocyte proliferative response to stimuli, and a malfunction of cell-mediated immunity at the molecular level. ESRD also increases patients' propensity for infections and malignancies as well as causing a diminished response to vaccination. Several factors influence the immunodeficiency in patients with ESRD, including uremic toxins, malnutrition, chronic inflammation, and the therapeutic dialysis modality. The alteration of T-cell function in ESRD has been considered to be a major factor underlying the impaired adaptive cellular immunity in these patients. However, cumulative evidence has suggested that the immune defect in ESRD can be caused by an Ag-presenting dendritic cell (DC) dysfunction in addition to a T-cell defect. It has been reported that ESRD has a deleterious effect on DCs both in terms of their number and function, although the precise mechanism by which DC function becomes altered in these patients is unclear. In this review, we discuss the effects of ESRD on the number and function of DCs and propose a possible molecular mechanism for DC dysfunction. We also address therapeutic approaches to improve immune function by optimally activating DCs in patients with ESRD.
Collapse
Affiliation(s)
- Ji Ung Kim
- Division of Nephrology, Department of Internal Medicine, Jeju National University Hospital, Jeju National University School of Medicine, Jeju 63241, Korea
| | - Miyeon Kim
- Division of Nephrology, Department of Internal Medicine, Jeju National University Hospital, Jeju National University School of Medicine, Jeju 63241, Korea
| | - Sinae Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea.,College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Tam Thanh Nguyen
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea.,College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Eunhye Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea.,College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Siyoung Lee
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea.,YbdYbiotech research center, Seoul 08589, Korea
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea.,College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea.,College of Veterinary Medicine, Veterinary Science Research Institute, Konkuk University, Seoul 05029, Korea
| | - Hyunwoo Kim
- Division of Nephrology, Department of Internal Medicine, Jeju National University Hospital, Jeju National University School of Medicine, Jeju 63241, Korea
| |
Collapse
|
23
|
Dixon KB, Davies SS, Kirabo A. Dendritic cells and isolevuglandins in immunity, inflammation, and hypertension. Am J Physiol Heart Circ Physiol 2016; 312:H368-H374. [PMID: 27986660 DOI: 10.1152/ajpheart.00603.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/02/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023]
Abstract
Hypertension is the major risk factor for morbidity and mortality from myocardial infarction, stroke, heart failure, and chronic kidney disease. Despite its importance, the pathogenesis of essential hypertension is poorly understood. During the past several years, it has become evident that T cells contribute to hypertension. Activated T cells accumulate in the perivascular space and the kidney and release cytokines that promote vascular dysfunction and end-organ damage. Although dendritic cells play a pivotal role in initiating adaptive immune responses, T cells have taken center stage in studies implicating the immune system in the genesis of hypertension. The mechanisms by which T cells are activated and the antigens involved are poorly understood. We recently showed that hypertension is associated with increased dendritic cell production of the TH17 polarizing cytokines, IL-6, IL-1β, and IL-23. This occurs in part by increased superoxide production via NADPH oxidase and protein modification by highly reactive isolevuglandins (IsoLGs). IsoLGs are produced via the isoprostane pathway of free radical-mediated lipid peroxidation and, when adducted to proteins, have the potential to act as neoantigens. In this review, we discuss recent advances in our understanding of the role of antigen-presenting dendritic cells in the pathophysiology of hypertension and highlight potential neoantigens that may contribute to this disease.
Collapse
Affiliation(s)
- Kala B Dixon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sean S Davies
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; and
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; .,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville Tennessee
| |
Collapse
|
24
|
Impact of Ivabradine on Inflammatory Markers in Chronic Heart Failure. J Immunol Res 2016; 2016:6949320. [PMID: 27822484 PMCID: PMC5086393 DOI: 10.1155/2016/6949320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/28/2016] [Accepted: 09/20/2016] [Indexed: 01/25/2023] Open
Abstract
Background. Inflammation plays a crucial role in the progression of chronic heart failure (CHF). Ivabradine is known to reduce the morbidity and mortality of patients with CHF under certain conditions. Beyond the reduction of heart rate, only limited knowledge exists about potential anti-inflammatory effects of ivabradine that might contribute to its benefit in CHF. Thus, the present study aimed to investigate the effect of ivabradine on systemic inflammation. Methods. In the present study, 33 patients with CHF due to dilated, ischemic, and hypertensive cardiomyopathy were treated with ivabradine according to the guidelines of the European Society of Cardiology (ESC). A number of circulating dendritic cells as well as inflammatory mediators were investigated using FACS analysis and ELISA, respectively, before and during ivabradine therapy. Results. Treatment with ivabradine resulted in a significant improvement of CHF symptoms as well as an increase in left ventricular ejection fraction. Moreover, ivabradine treatment led to a significant reduction of TNF-alpha (TNF-α) serum levels and a reconstitution of circulating dendritic cells which are known to be reduced in patients with CHF. Conclusion. We show that treatment with ivabradine in patients with CHF resulted in an improvement of HF symptoms and ejection fraction as well as a normalization of inflammatory mediators.
Collapse
|
25
|
Rohm I, Aderhold N, Ratka J, Goebel B, Franz M, Pistulli R, Gecks T, Figulla HR, Yilmaz A, Jung C. Hypobaric hypoxia in 3000 m altitude leads to a significant decrease in circulating plasmacytoid dendritic cells in humans. Clin Hemorheol Microcirc 2016; 63:257-65. [DOI: 10.3233/ch-152035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
26
|
Paul K, Franke S, Nadal J, Schmid M, Yilmaz A, Kretzschmar D, Bärthlein B, Titze S, Koettgen A, Wolf G, Busch M. Inflammation, vitamin D and dendritic cell precursors in chronic kidney disease. Clin Exp Immunol 2016; 186:86-95. [PMID: 27414487 DOI: 10.1111/cei.12844] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2016] [Indexed: 01/14/2023] Open
Abstract
Decreased blood dendritic cell precursors (DCP) count is linked with atherosclerotic disease, while reduction of circulating DCP is also seen in patients with chronic kidney disease (CKD). As poor vitamin D status could be linked to a compromised innate immune response, we hypothesized that vitamin D status might be involved in the decrease in circulating DCP in CKD. Moreover, the potential role of inflammation was considered. Circulating myeloid (mDCP), plasmacytoid (pDCP) and total DCP (tDCP) were analysed using flow cytometry in 287 patients with CKD stage 3. Serum 25(OH)D and 1,25(OH)2D levels were measured using enzyme-linked immunosorbent assays (ELISA), interleukin (IL)-6, IL-10 and tumour necrosis factor (TNF)-α using cytometric bead array, C-reactive protein (CRP) using a high-sensitivity (hs) ELISA. Contrary to our hypothesis, there was no association between vitamin D levels and DCP, although their number was decreased significantly in CKD (P < 0·001). Instead, mDCP (r = -0·211) and tDCP (r = -0·188,) were associated slightly negatively with hsCRP but positively with the estimated glomerular filtration rate (eGFR, r = 0·314 for tDCP). According to multivariate linear regression, only higher hsCRP concentration and the presence of diabetes mellitus had a significant negative influence on DCP count (P < 0·03, respectively) but not vitamin D, age and eGFR. A significant impact of vitamin D on the reduction of circulating DCP in CKD 3 patients can be neglected. Instead, inflammation as a common phenomenon in CKD and diabetes mellitus had the main influence on the decrease in DCP. Thus, a potential role for DCP as a sensitive marker of inflammation and cardiovascular risk should be elucidated in future studies.
Collapse
Affiliation(s)
- K Paul
- Department of Internal Medicine III, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - S Franke
- Department of Internal Medicine III, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - J Nadal
- Institute of Medical Biometry, Informatics and Epidemiology at Rhenish Friedrich-Wilhelm University, Bonn, Germany
| | - M Schmid
- Institute of Medical Biometry, Informatics and Epidemiology at Rhenish Friedrich-Wilhelm University, Bonn, Germany
| | - A Yilmaz
- Department of Internal Medicine II, Division of Cardiology, Elisabeth Klinikum Schmalkalden GmbH, Schmalkalden, Germany
| | - D Kretzschmar
- Department of Internal Medicine I, Division of Cardiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - B Bärthlein
- Chair of Medical Informatics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - S Titze
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
| | - A Koettgen
- Department of Medicine IV, University Hospital Freiburg, Freiburg, Germany
| | - G Wolf
- Department of Internal Medicine III, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - M Busch
- Department of Internal Medicine III, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | | |
Collapse
|
27
|
Dendritic Cells and Their Role in Cardiovascular Diseases: A View on Human Studies. J Immunol Res 2016; 2016:5946807. [PMID: 27088098 PMCID: PMC4818818 DOI: 10.1155/2016/5946807] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/22/2016] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
The antigen-presenting dendritic cells (DCs) are key to the immunological response, with different functions ascribed ranging from cellular immune activation to induction of tolerance. Such immunological responses are involved in the pathophysiological mechanisms of cardiovascular diseases, with DCs shown to play a role in atherosclerosis, hypertension, and heart failure and most notably following heart transplantation. A better understanding of the interplay between the immune system and cardiovascular diseases will therefore be critical for developing novel therapeutic treatments as well as innovative monitoring tools for disease progression. As such, the present review will provide an overview of DCs involvement in the pathophysiology of cardiovascular diseases and how targeting these cells may have beneficial effects for the prognosis of patients.
Collapse
|
28
|
Yilmaz A, Ratka J, Rohm I, Pistulli R, Goebel B, Asadi Y, Petri A, Kiehntopf M, Figulla HR, Jung C. Decrease in circulating plasmacytoid dendritic cells during short-term systemic normobaric hypoxia. Eur J Clin Invest 2016; 46:115-22. [PMID: 25652640 DOI: 10.1111/eci.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/31/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND During exposure to high altitude, the immune system is altered. During hypoxia, an increase in interleukin (IL)-6 and high sensitivity C-reactive protein (hs-CRP), and an increase in natural killer cells and decrease in T cells in blood was shown. However, the impact of hypoxia on dendritic cells has not been investigated yet. MATERIAL AND METHODS Twelve healthy volunteers were subjected to a transient normobaric hypoxia for 6·5 h simulating an oxygen concentration at 5500 m. During exposure to hypoxia, blood samples were collected and analysed by flow cytometrical cell sorting (FACS) for circulating myeloid (mDCs) and plasmacytoid (pDCs) DCs. Serum levels of IL-6 and tumour necrosis factor (TNF)-α were analysed. In a cell culture hypoxia chamber, blood samples were subjected to the same hypoxia and analysed regarding DCs. RESULTS Exposure to normobaric hypoxia induced a significant decrease in circulating pDCs about 45% (P = 0·001) but not of mDC compared to baseline normoxia. Furthermore, we observed a significant increase of TNF-α about 340% (P = 0·03) and of IL-6 about 286% (P = 0·002). In cell culture experiments exposure of blood to hypoxia led to no significant changes in DCs, so that a direct cytotoxic effect was excluded. During hypoxia, we observed a transient increase in stromal-derived factor 1 (SDF-1) which is important for pDC tissue recruitment. CONCLUSIONS We show a significant decrease in circulating pDCs during hypoxia in parallel to a pro-inflammatory response. Further studies are necessary to evaluate whether the decrease in circulating pDCs might be the result of an enhanced tissue recruitment.
Collapse
Affiliation(s)
- Atilla Yilmaz
- Internal Medicine II, Elisabeth Hospital Schmalkalden, Schmalkalden, Germany
| | - Josi Ratka
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Ilonka Rohm
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Rudin Pistulli
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Bjorn Goebel
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Yahya Asadi
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Alexander Petri
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Michael Kiehntopf
- Department of Clinical Chemistry & Laboratory Medicine, University Hospital of Jena, Jena, Germany
| | - Hans R Figulla
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Christian Jung
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| |
Collapse
|
29
|
Systemic inflammatory response following acute myocardial infarction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2015; 12:305-12. [PMID: 26089856 PMCID: PMC4460175 DOI: 10.11909/j.issn.1671-5411.2015.03.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/02/2015] [Accepted: 04/11/2015] [Indexed: 12/31/2022]
Abstract
Acute cardiomyocyte necrosis in the infarcted heart generates damage-associated molecular patterns, activating complement and toll-like receptor/interleukin-1 signaling, and triggering an intense inflammatory response. Inflammasomes also recognize danger signals and mediate sterile inflammatory response following acute myocardial infarction (AMI). Inflammatory response serves to repair the heart, but excessive inflammation leads to adverse left ventricular remodeling and heart failure. In addition to local inflammation, profound systemic inflammation response has been documented in patients with AMI, which includes elevation of circulating inflammatory cytokines, chemokines and cell adhesion molecules, and activation of peripheral leukocytes and platelets. The excessive inflammatory response could be caused by a deregulated immune system. AMI is also associated with bone marrow activation and spleen monocytopoiesis, which sustains a continuous supply of monocytes at the site of inflammation. Accumulating evidence has shown that systemic inflammation aggravates atherosclerosis and markers for systemic inflammation are predictors of adverse clinical outcomes (such as death, recurrent myocardial infarction, and heart failure) in patients with AMI.
Collapse
|
30
|
Decrease in circulating dendritic cell precursors in patients with peripheral artery disease. Mediators Inflamm 2015; 2015:450957. [PMID: 25960616 PMCID: PMC4413958 DOI: 10.1155/2015/450957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/10/2015] [Indexed: 12/23/2022] Open
Abstract
Peripheral artery disease (PAD) is a common manifestation of atherosclerosis. Inflammation is important for initiation and progression of the disease. Dendritic cells (DCs) as antigen-presenting cells play an important role in the immune system. Therefore, we hypothesize that, in patients with PAD, DCPs might be reduced in blood due to their recruitment into the vascular wall and induce a proinflammatory response. The numbers of myeloid DCPs, plasmacytoid DCPs, and total DCPs were analyzed by flow cytometry in blood of patients with PAD (n = 52) compared to controls (n = 60). Femoralis plaques (n = 12) of patients who underwent surgery were immunostained for CD209 and CD83 (mDCs) as well as CD304, CD123 (pDCs), and HLA-DR. In patients with PAD, a significant decrease in mDCPs, pDCPs, and tDCPs was observed. In immunostaining, markers indicative for mDCs (CD209: 16 versus 8 cells/0.1 mm(2), P = 0.02; CD83: 19 versus 5 cells/0.1 mm(2), P = 0.03) were significantly elevated in femoralis plaques compared to control vessels. We show for the first time that mDCPs, pDCPs, and tDCPs are significantly reduced in patients with PAD. Immunohistochemical analysis unraveled that the decrease in DCPs might be due to their recruitment into atherosclerotic plaques.
Collapse
|
31
|
Seropian IM, Sonnino C, Van Tassell BW, Biasucci LM, Abbate A. Inflammatory markers in ST-elevation acute myocardial infarction. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2015; 5:382-95. [PMID: 25681486 DOI: 10.1177/2048872615568965] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 01/02/2015] [Indexed: 01/05/2023]
Abstract
After acute myocardial infarction, ventricular remodeling is characterized by changes at the molecular, structural, geometrical and functional level that determine progression to heart failure. Inflammation plays a key role in wound healing and scar formation, affecting ventricular remodeling. Several, rather different, components of the inflammatory response were studied as biomarkers in ST-elevation acute myocardial infarction. Widely available and inexpensive tests, such as leukocyte count at admission, as well as more sophisticated immunoassays provide powerful predictors of adverse outcome in patients with ST-elevation acute myocardial infarction. We review the value of inflammatory markers in ST-elevation acute myocardial infarction and their association with ventricular remodeling, heart failure and sudden death. In conclusion, the use of these biomarkers may identify subjects at greater risk of adverse events and perhaps provide an insight into the mechanisms of disease progression.
Collapse
Affiliation(s)
- Ignacio M Seropian
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Chiara Sonnino
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA Department of Cardiovascular Medicine, Catholic University, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA School of Pharmacy, Virginia Commonwealth University, USA
| | - Luigi M Biasucci
- Department of Cardiovascular Medicine, Catholic University, Italy
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA
| |
Collapse
|
32
|
Zernecke A. Dendritic cells in atherosclerosis: evidence in mice and humans. Arterioscler Thromb Vasc Biol 2015; 35:763-70. [PMID: 25675999 DOI: 10.1161/atvbaha.114.303566] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Atherosclerotic vascular disease is driven by chronic inflammation involving both innate and adaptive immune responses. Dendritic cells (DCs) are found in healthy arteries and accumulate in atherosclerotic lesions and engage in diverse pathogenic and protective mechanisms during atherogenesis. DCs contribute to early foam cell formation, regulate lipid metabolism, and control pro- and antiatherosclerotic T-cell responses by multifarious mechanisms. We, here, review the roles of DCs and plasmacytoid DCs in experimental models of atherosclerosis and the approaches to target DCs in therapeutic vaccination strategies. We, furthermore, discuss the evidence of the potential function of DCs in human atherosclerosis, and dissect the efforts to harness DC subsets as biomarkers of disease. Finally, we discuss necessary future steps that will help to understand the specific contribution of bona fide DCs in atherosclerosis to move toward novel therapeutic approaches.
Collapse
Affiliation(s)
- Alma Zernecke
- From the Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
33
|
Yu K, Dong Q, Mao X, Meng K, Zhao X, Ji Q, Wu B, Zhong Y, Zhu Z, Liu Y, Zhang W, Tony H, Shi H, Zeng Q. Disruption of the TSLP-TSLPR-LAP signaling between epithelial and dendritic cells through hyperlipidemia contributes to regulatory T-Cell defects in atherosclerotic mice. Atherosclerosis 2014; 238:278-88. [PMID: 25544178 DOI: 10.1016/j.atherosclerosis.2014.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/08/2023]
Abstract
Regulatory T-Cells (Tregs) play a protective role against the development of atherosclerosis. Moreover, thymic stromal lymphopoietin (TSLP)/thymic stromal lymphopoietin receptor (TSLPR) signaling in myeloid dendritic cells (DCs) promote Treg differentiation. Here, we examined the potential role of TSLP/TSLPR on Treg homeostasis in atherosclerosis. The frequencies of both latency-associated peptide (LAP)(+) and Foxp3(+) Tregs were reduced in the thymus and spleen of ApoE(-/-) mice compared with C57BL/6 mice, and this effect was associated with decreased thymic output. The tolerogenic function of DCs obtained from ApoE(-/-) mice was compromised compared with those from C57BL/6 mice. The expression of TSLP and TSLPR was also inhibited in ApoE(-/-) mice. In addition, we found that ox-LDL attenuated TSLP expression in cultured thymic epithelial cells (TECs) through the activation of retinoid X receptor alpha (RXRA) and IL-1β and decreased LAP and PD-L1 expression in oxLDL-activated DCs while both were up-regulated in TSLP-activated DCs. We also observed that the TSLP-DCs mediated differentiation of Tregs was abrogated through LAP neutralization. Furthermore, TSLP injection rescued Treg defects in ApoE(-/-) mice. These findings suggest that Treg defects in ApoE(-/-) mice might partially be attributed to the disruption of TSLP-TSLPR-LAP signaling in epithelial cells (ECs) and DCs.
Collapse
Affiliation(s)
- Kunwu Yu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Dong
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Mao
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Meng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Zhao
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingwei Ji
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Bangwei Wu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yucheng Zhong
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Zhu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhou Liu
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hasahya Tony
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Huairui Shi
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
34
|
Nagai T, Honda S, Sugano Y, Matsuyama TA, Ohta-Ogo K, Asaumi Y, Ikeda Y, Kusano K, Ishihara M, Yasuda S, Ogawa H, Ishibashi-Ueda H, Anzai T. Decreased myocardial dendritic cells is associated with impaired reparative fibrosis and development of cardiac rupture after myocardial infarction in humans. J Am Heart Assoc 2014; 3:e000839. [PMID: 24895162 PMCID: PMC4309075 DOI: 10.1161/jaha.114.000839] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Dendritic cells (DC) play pivotal roles in regulating the immune system and inflammatory response. We previously reported DC infiltration in the infarcted heart and its immunoprotective roles in the post‐infarction healing process after experimental myocardial infarction (MI). However, its clinical significance has not been determined. Methods and Results The degree of DC infiltration and its correlation with the post‐infarction healing process in the human infarcted heart were investigated in 24 autopsy subjects after ST‐elevation MI. Patients were divided into two groups according to the presence (n=13) or absence (n=11) of cardiac rupture. The numbers of infiltrated DC and macrophages and the extent of fibrosis in the infarcted area were examined. In the rupture group, CD68+ macrophage infiltration was increased and CD209+ DC, and CD11c+ DC infiltration and the extent of reparative fibrosis were decreased compared with the non‐rupture group, under matched baseline characteristics including the time from onset to death and use of revascularization. Furthermore, there was a significant positive correlation between the number of infiltrating CD209+ DC, and CD11c+ DC and the extent of reparative fibrosis. Conclusions Decreased number of DC in human‐infarcted myocardial tissue was associated with increased macrophage infiltration, impaired reparative fibrosis, and the development of cardiac rupture after MI. These findings suggest a protective role of DC in post‐MI inflammation and the subsequent healing process.
Collapse
Affiliation(s)
- Toshiyuki Nagai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Satoshi Honda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Yasuo Sugano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Taka-aki Matsuyama
- Department of Clinical Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan (T.M., K.O.O., Y.I., H.I.U.)
| | - Keiko Ohta-Ogo
- Department of Clinical Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan (T.M., K.O.O., Y.I., H.I.U.)
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Yoshihiko Ikeda
- Department of Clinical Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan (T.M., K.O.O., Y.I., H.I.U.)
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Masaharu Ishihara
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.) Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (H.O.)
| | - Hatsue Ishibashi-Ueda
- Department of Clinical Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan (T.M., K.O.O., Y.I., H.I.U.)
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (T.N., S.H., Y.S., Y.A., K.K., M.I., S.Y., H.O., T.A.)
| |
Collapse
|
35
|
Chistiakov DA, Sobenin IA, Orekhov AN, Bobryshev YV. Dendritic cells in atherosclerotic inflammation: the complexity of functions and the peculiarities of pathophysiological effects. Front Physiol 2014; 5:196. [PMID: 24904430 PMCID: PMC4034414 DOI: 10.3389/fphys.2014.00196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/09/2014] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is considered as a chronic disease of arterial wall, with a strong contribution of inflammation. Dendritic cells (DCs) play a crucial role in the initiation of proatherogenic inflammatory response. Mature DCs present self-antigens thereby supporting differentiation of naïve T cells to effector cells that further propagate atherosclerotic inflammation. Regulatory T cells (Tregs) can suppress proinflammatory function of mature DCs. In contrast, immature DCs are able to induce Tregs and prevent differentiation of naïve T cells to proinflammatory effector T cells by initiating apoptosis and anergy in naïve T cells. Indeed, immature DCs showed tolerogenic and anti-inflammatory properties. Thus, DCs play a double role in atherosclerosis: mature DCs are proatherogenic while immature DCs appear to be anti-atherogenic. Tolerogenic and anti-inflammatory capacity of immature DCs can be therefore utilized for the development of new immunotherapeutic strategies against atherosclerosis.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical University Moscow, Russia
| | - Igor A Sobenin
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Russia ; Institute of General Pathology and Pathophysiology, Russian Academy of Sciences Moscow, Russia ; Laboratory of Medical Genetics, Russian Cardiology Research and Production Complex Moscow, Russia
| | - Alexander N Orekhov
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Russia ; Institute of General Pathology and Pathophysiology, Russian Academy of Sciences Moscow, Russia
| | - Yuri V Bobryshev
- Skolkovo Innovative Center, Institute for Atherosclerosis Research Moscow, Russia ; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Kensington, Sydney NSW, Australia
| |
Collapse
|
36
|
Affiliation(s)
- Anette Christ
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands (A.C., L.T., B.L., E.A.L.B.); Department of Cell Biology, Institute for Biomedical Engineering, Aachen University Hospital, Aachen, Germany (A.C.); and Department of Pathology, Amsterdam Medical Center, Amsterdam, The Netherlands (M.J.A.P.D.)
| | | | | | | | | |
Collapse
|
37
|
Paul K, Kretzschmar D, Yilmaz A, Bärthlein B, Titze S, Wolf G, Busch M. Circulating dendritic cell precursors in chronic kidney disease: a cross-sectional study. BMC Nephrol 2013; 14:274. [PMID: 24325304 PMCID: PMC3878881 DOI: 10.1186/1471-2369-14-274] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 11/25/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Dendritic cells (DC) are professional antigen-presenting cells in the immune system. They patrol the blood as circulating dendritic cell precursors (DCP). Decreased blood DCP count has been shown to be related to atherosclerotic plaque burden. Since chronic kidney disease (CKD) is associated with chronic inflammation and increased cardiovascular risk, the aim of our study was to investigate a potential effect of CKD on circulating DCP numbers especially in patients with a history of cardiovascular disease. METHODS The number of circulating myeloid (mDCP), plasmacytoid (pDCP), and total DCP (tDCP) was analysed by flow cytometry in 245 patients with CKD stage 3 (with and without known cardiovascular events) and 85 coronary healthy controls. In addition, data were compared with a historical group of 130 patients with known coronary artery disease (CAD). RESULTS Compared to controls, patients with CKD 3 revealed a significant decrease in circulating mDCP (-29%), pDCP (-43%), and tDCP (-38%) (P < 0.001, respectively). Compared with CAD-patients, the decrease in circulating DCP in CKD was comparable or even more pronounced indicating a potential role for DCP in cardiovascular risk potentiation due to CKD. CONCLUSIONS Based on previous findings in CAD, the marked decrease of DCP in CKD implicates a potential role for DCP as a mediator of cardiovascular disease. Whether DCP in CKD may act as new cardiovascular biomarkers needs to be established in future prospective trials.
Collapse
Affiliation(s)
- Katharina Paul
- Department of Internal Medicine III, Division of Nephrology, Jena University Hospital Friedrich-Schiller University, Erlanger Allee 101, Jena, 07740 Germany.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall characterized by innate and adaptive immune system involvement. A key component of atherosclerotic plaque inflammation is the persistence of different innate immune cell types including mast cells, neutrophils, natural killer cells, monocytes, macrophages and dendritic cells. Several endogenous signals such as oxidized low-density lipoproteins, and exogenous signals such as lipopolysaccharides, trigger the activation of these cells. In particular, these signals orchestrate the early and late inflammatory responses through the secretion of pro-inflammatory cytokines and contribute to plaque evolution through the formation of foam cells, among other events. In this review we discuss how innate immune system cells affect atherosclerosis pathogenesis.
Collapse
|
39
|
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
|
40
|
Bobryshev YV, Karagodin VP, Orekhov AN. Dendritic cells and their role in immune reactions of atherosclerosis. ACTA ACUST UNITED AC 2013. [DOI: 10.1134/s1990519x1302003x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
41
|
Chistiakov DA, Sobenin IA, Orekhov AN, Bobryshev YV. WITHDRAWN: Dendritic cells: A double-edge sword in atherosclerotic inflammation. Atherosclerosis 2013:S0021-9150(13)00190-1. [PMID: 23578357 DOI: 10.1016/j.atherosclerosis.2013.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/07/2013] [Accepted: 03/09/2013] [Indexed: 11/25/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Pirogov Russian State Medical University, Department of Medical Nanobiotechnology, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | | | | | | |
Collapse
|
42
|
Bisoprolol reverses epinephrine-mediated inhibition of cell emigration through increases in the expression of β-arrestin 2 and CCR7 and PI3K phosphorylation, in dendritic cells loaded with cholesterol. Thromb Res 2013; 131:230-7. [PMID: 23290307 DOI: 10.1016/j.thromres.2012.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 01/09/2023]
Abstract
The effect of bisoprolol on dendritic cell (DC) migration was investigated, including the analysis of protein expression, cytokine secretion and activation of the PI3K-pathway. The chemotactic cell numbers in cholesterol-loaded DCs treated with epinephrine were significantly decreased by 26.66±6.29% (6h), 35.67±2.91% (12h) and 29.33±1.09% (24h). This effect was significantly reversed by 46.00±10.65% (6h), 64.25±6.77% (12h) and 55.74±5.51% (24h) when bisoprolol and epinephrine were both present. In cholesterol-loaded DCs, treatment with epinephrine significantly increased AR-β1 protein expression by 56.99±4.87%, but inhibited β-arrestin 2 and CCR7 protein expression by 30.51±4.22% and 25.31±0.04%, respectively. These effects were reversed by bisoprolol by 36.87±4.40%, 41.47±3.95% and 30.14±0.54%, respectively. TNF-α and MMP9 levels were decreased by 68.33±4.00% and 39.57±9.21% in cholesterol-loaded DCs treated with epinephrine. In contrast, when bisoprolol and epinephrine were administered together, the secretion of these proteins was significantly increased by 233.81±37.06 % and 76.66±14.21%, respectively. Treatment with epinephrine decreased PI3K-phosphorylation by 31.88±2.79%, 40.24±5.69% and 30.93±4.66% at 15, 30 and 60min, respectively, whereas the effect of epinephrine on the expression of phosphorylated PI3K was reversed by 49.49±12.12%, 70.93±16.14% and 47.62±6.00%, respectively, when cells were treated with both bisoprolol and epinephrine. Wortmannin inhibited the effects of bisoprolol on PI3K-phosphorylation (38.63±6.12%), the expression of CCR7 (23.4±2.72%), the secretion of TNF-α (69.46±4.48%) and MMP9 (43.15±4.63%), and the number of chemotactic cells (36.84±5.22%). This is the first study to establish a signaling pathway, epinephrine-AR-β1-β-arrestin2-PI3K-MMP9/CCR7, which plays a critical role in the migration of DCs.
Collapse
|
43
|
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
|
44
|
Businaro R. Neuroimmunology of the atherosclerotic plaque: a morphological approach. J Neuroimmune Pharmacol 2012; 8:15-27. [PMID: 23150034 DOI: 10.1007/s11481-012-9421-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 11/05/2012] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is a chronic inflammatory process, lasting for several decades until the onset of its clinical manifestations. The progression of the atherosclerotic lesion to a stable fibrotic plaque, narrowing the vascular lumen, or to a vulnerable plaque leading to main vascular complications, is associated to the involvement of several cell subpopulations of the innate as well as of the adaptive immunity, and to the release of chemokines and pro-inflammatory cytokines. Emerging evidence outlines that the cardiovascular risk is dependent on stress-mediators influencing cell migration and vascular remodeling. The view that atherosclerosis is initiated by monocytes and lymphocytes adhering to dysfunctional endothelial cells is substantiated by experimental and clinical observations. Macrophages, dendritic cells, T and B lymphocytes, granulocytes accumulating into the subendothelial space secrete and are stimulated by soluble factors, including peptides, proteases and cytokines acting synergistically. The final step of the disease, leading to plaque destabilization and rupture, is induced by the release, at the level of the fibrous cap, of metalloproteinases and elastases by the activated leukocytes which accumulate locally. Recruitment of specific cell subpopulations as well as the progression of atherosclerotic lesions towards a stable or an unstable phenotype, are related to the unbalance between pro-atherogenic and anti-atherogenic factors. In this connection stress hormones deserve particular attention, since their role in vascular remodeling, via vascular smooth cell proliferation, as well as in neoangiogenesis, via stimulation of endothelial cell proliferation and migration, has been already established.
Collapse
Affiliation(s)
- Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy.
| |
Collapse
|
45
|
Dopheide JF, Obst V, Doppler C, Radmacher MC, Scheer M, Radsak MP, Gori T, Warnholtz A, Fottner C, Daiber A, Münzel T, Espinola-Klein C. Phenotypic characterisation of pro-inflammatory monocytes and dendritic cells in peripheral arterial disease. Thromb Haemost 2012; 108:1198-207. [PMID: 23093299 DOI: 10.1160/th12-05-0327] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 09/11/2012] [Indexed: 11/05/2022]
Abstract
Atherosclerosis is a chronic inflammatory process involving antigen-presenting cells like monocytes and dendritic cells (DC). The aim of this study was to perform a phenotypic characterisation of these cell types in patients with different degrees of peripheral arterial disease (PAD). Sixty patients with PAD [N= 30 intermittent claudication (IC), N= 30 critical limb ischemia (CLI)] and 30 controls were included. Peripheral blood leucocytes were analysed from peripheral blood by flow cytometry using different gating strategies to directly identify and analyse monocytes, myeloid DC, (mDC) and plasmacytoid DC (pDC). PAD patients showed a significantly higher proportion of proinflammatory CD14++CD16+ monocytes (p<0.0001) compared with healthy individuals. We found an increased number of mDC/ml and a reduced number of pDC/ml (both p<0.01) in PAD patients, leading to a shift in the mDC/pDC ratio (p<0.01). As compared to patients with intermittent claudication, CLI patients presented a reduced expression of HLA-DR (p<0.01), CD86 and CD40 on both mDCs and pDCs (p<0.01). Peripheral blood monocytes show a proinflammatory phenotype in PAD patients compared to controls. In contrast, CLI patients show a reduced expression of proinflammatory markers. We hypothesise that severe ischaemia and/or prolonged inflammation in CLI might lead to a paradoxical attenuation in the proinflammatory membrane pattern of circulating mononuclear cells, possibly hindering an adequate regulatory function of mDCs and pDCs and favouring the progression of disease.
Collapse
Affiliation(s)
- Jörn F Dopheide
- Department of Internal Medicine II, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Grassia G, MacRitchie N, Platt AM, Brewer JM, Garside P, Maffia P. Plasmacytoid dendritic cells: biomarkers or potential therapeutic targets in atherosclerosis? Pharmacol Ther 2012; 137:172-82. [PMID: 23059425 DOI: 10.1016/j.pharmthera.2012.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 09/21/2012] [Indexed: 12/28/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) represent a unique subset of dendritic cells that play distinct and critical roles in the immune response. Importantly, pDCs play a pivotal role in several chronic autoimmune diseases strongly characterized by an increased risk of vascular pathology. Clinical studies have shown that pDCs are detectable in atherosclerotic plaques and others have suggested an association between reduced numbers of circulating pDCs and cardiovascular events. Although the causal relationship between pDCs and atherosclerosis is still uncertain, recent results from mouse models are starting to define the specific role(s) of pDCs in the disease process. In this review, we will discuss the role of pDCs in innate and adaptive immunity, the emerging evidence demonstrating the contribution of pDCs to vascular pathology and we will consider the possible impact of pDCs on the acceleration of atherosclerosis in chronic inflammatory autoimmune diseases. Finally, we will discuss how pDCs could be targeted for therapeutic utility.
Collapse
Affiliation(s)
- Gianluca Grassia
- Department of Experimental Pharmacology, University of Naples Federico II, 80131 Naples, Italy
| | | | | | | | | | | |
Collapse
|
47
|
Matusik P, Guzik B, Weber C, Guzik TJ. Do we know enough about the immune pathogenesis of acute coronary syndromes to improve clinical practice? Thromb Haemost 2012; 108:443-56. [PMID: 22872109 DOI: 10.1160/th12-05-0341] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/17/2012] [Indexed: 11/05/2022]
Abstract
Morbidities related to atherosclerosis, such as acute coronary syndromes (ACS) including unstable angina and myocardial infarction, remain leading causes of mortality. Unstable plaques are inflamed and infiltrated with macrophages and T lymphocytes. Activated dendritic cells interact with T cells, yielding predominantly Th1 responses involving interferon-gamma (IFN-γ) and tumour necrosis factor-alpha (TNF-α), while the role of interleukin 17 (IL-17) is questionable. The expansion of CD28nullCD4 or CD8 T cells as well as pattern recognition receptors activation (especially Toll-like receptors; TLR2 and TLR4) is characteristic for unstable plaque. Inflammation modifies platelet and fibrin clot characteristics, which are critical for ACS. Understanding of the inflammatory mechanisms of atherothrombosis, bridging inflammation, oxidative stress and immune regulation, will allow for the detection of subjects at risk, through the use of novel biomarkers and imaging techniques including intravascular ultrasound, molecular targeting, magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET). Moreover, understanding the specific inflammatory pathways of plaque rupture and atherothrombosis may allow for immunomodulation of ACS. Statins and anti-platelet drugs are anti-inflammatory, but importance of immune events in ACS warrants the introduction of novel, specific treatments directed either on cytokines, TLRs or inflammasomes. While the prime time for the introduction of immunologically inspired diagnostic tests and treatments for atherosclerosis have not come yet, we are closer than ever before to finally being able to benefit from this vast body of experimental and clinical evidence. This paper provides a comprehensive review of the role of the immune system and inflammation in ACS.
Collapse
Affiliation(s)
- Pawel Matusik
- Translational Medicine Laboratory, Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Kracow, Poland
| | | | | | | |
Collapse
|
48
|
Fukui D, Yasukawa H, Sugi Y, Oba T, Nagata T, Kyogoku S, Futamata N, Yokoyama T, Yokoyama S, Kai H, Ueno T, Kage M, Imaizumi T. Transient reduction and activation of circulating dendritic cells in patients with acute myocardial infarction. Int J Cardiol 2012; 160:216-9. [PMID: 22841473 DOI: 10.1016/j.ijcard.2012.06.070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/09/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Dendritic cells (DCs) are highly potent professional antigen-presenting cells that play a central role in initiating the primary immune response. Accumulating evidence suggests that immune-mediated inflammation plays an important role in the pathophysiology of AMI, but the mechanism that triggers such immune responses is unknown. METHODS Using multi-color flow-cytometry, we determined the numbers of circulating myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) in patients with AMI (n = 26) or stable angina pectoris (SAP) (n = 19), and in age-matched control subjects (n = 19). The DC activation markers CD40 and CD83 were also measured. RESULTS On admission, circulating mDC and pDC counts were significantly lower in AMI patients compared to control subjects and SAP patients (mDC, P < 0.01; pDC, P < 0.05). The activation markers of mDCs in AMI patients were significantly higher and returned to the levels of control subjects or SAP patients 3 days after AMI (mDC, P < 0.05; pDC, P < 0.05). Reductions of circulating mDC and pDC numbers were restored 7 days after the onset of AMI. Furthermore, we found that the recovery of the circulating DC numbers 14 days after AMI was correlated with the alterations of creatine kinase-MB (CK-MB) (mDC, r = 0.48, P < 0.05; pDC, r=0.52, P < 0.01) and brain natriuretic peptide (BNP) (mDC, r = 0.53, P < 0.01; pDC, r = 0.51, P < 0.01). CONCLUSION Our findings suggest that the transient reduction and activation of circulating DCs may play important roles in the pathophysiology of myocardial injury after AMI.
Collapse
Affiliation(s)
- Daisuke Fukui
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Feig JE, Feig JL. Macrophages, dendritic cells, and regression of atherosclerosis. Front Physiol 2012; 3:286. [PMID: 22934038 PMCID: PMC3429058 DOI: 10.3389/fphys.2012.00286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/29/2012] [Indexed: 01/15/2023] Open
Abstract
Atherosclerosis is the number one cause of death in the Western world. It results from the interaction between modified lipoproteins and cells such as macrophages, dendritic cells (DCs), T cells, and other cellular elements present in the arterial wall. This inflammatory process can ultimately lead to the development of complex lesions, or plaques, that protrude into the arterial lumen. Ultimately, plaque rupture and thrombosis can occur leading to the clinical complications of myocardial infarction or stroke. Although each of the cell types plays roles in the pathogenesis of atherosclerosis, the focus of this review will be primarily on the macrophages and DCs. The role of these two cell types in atherosclerosis is discussed, with a particular emphasis on their involvement in atherosclerosis regression.
Collapse
Affiliation(s)
- Jonathan E Feig
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai Medical Center NY, USA
| | | |
Collapse
|
50
|
Elevated circulating interleukin-27 in patients with coronary artery disease is associated with dendritic cells, oxidized low-density lipoprotein, and severity of coronary artery stenosis. Mediators Inflamm 2012; 2012:506283. [PMID: 22911112 PMCID: PMC3403490 DOI: 10.1155/2012/506283] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/26/2012] [Accepted: 05/15/2012] [Indexed: 12/21/2022] Open
Abstract
Coronary artery disease (CAD) is an immune-mediated chronic inflammatory disease mainly caused by atherosclerosis. The aims of this study were to investigate the role of interleukin-27 (IL-27) in patients with CAD and the severity of coronary artery lesions, which was evaluated by Gensini score and to investigate the biosynthesis of IL-27 and oxidized low-density lipoprotein (ox-LDL) in vitro using monocyte-derived dendritic cells (DCs). To this aim, plasma levels of IL-27, ox-LDL, and Gensini score were analyzed in patients with CAD (n = 136) and normal subjects (controls, n = 29). IL-27 concentration of the supernatant and the mRNA expression levels of p28 and ebi3, subunits of IL-27, from cultured immature DCs incubated with different concentrations of ox-LDL for 24 h were also analyzed. We found that circulating IL-27 levels were significantly elevated in patients with CAD than in controls (P < 0.01), and positively correlated to ox-LDL and Gensini score. ox-LDL dose-dependently upregulated expression of both IL-27 protein and IL-27 (p28 and EBI3) mRNA in vitro, indicating that ox-LDL can stimulate DCs to produce IL-27. These results demonstrate that IL-27 might regulate the network of immunity and inflammation in the pathogenesis of atherosclerosis.
Collapse
|