1
|
Askarizadeh F, Karav S, Jamialahmadi T, Sahebkar A. Impact of statin therapy on CD40:CD40L signaling: mechanistic insights and therapeutic opportunities. Pharmacol Rep 2025; 77:43-71. [PMID: 39680334 DOI: 10.1007/s43440-024-00678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024]
Abstract
Statins are widely utilized to reduce cholesterol levels, particularly in cardiovascular diseases. They interface with cholesterol synthesis by inhibiting the 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase enzyme. Besides their primary effect, statins demonstrate anti-inflammatory and immune-modulating properties in various diseases, highlighting the pleiotropic effect of these drugs. The CD40:CD40L signaling pathway is considered a prominent inflammatory pathway in multiple diseases, including autoimmune, inflammatory, and cardiovascular diseases. The findings from clinical trials and in vitro and in vivo studies suggest the potential anti-inflammatory effect of statins in modulating the CD40 signaling pathway and downstream inflammatory mediator. Accordingly, as its classic ligand, statins can suppress immune responses in autoimmune diseases by inhibiting CD40 expression and blocking its interaction with CD40L. Additionally, statins affect intracellular signaling and inhibit inflammatory mediator secretion in chronic inflammatory diseases like asthma and autoimmune disorders such as myasthenia gravis, multiple sclerosis, systemic lupus erymanthus, and cardiovascular diseases like atherosclerosis. However, it is essential to note that the anti-inflammatory effect of statins may vary depending on the specific type of statin used. In this study, we aim to explore the potential anti-inflammatory effects of statins in treating inflammatory diseases by examining their role in regulating immune responses, particularly their impact on the CD40:CD40L signaling pathway, through a comprehensive review of existing literature.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [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: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
3
|
Cui Q, Li W, Wang D, Wang S, Liu A, Zhang G, Yang Y, Ge T, He G, Yu J. Immune signature and phagocytosis of circulating DC subsets in healthy adults during aging. Int Immunopharmacol 2024; 130:111715. [PMID: 38382263 DOI: 10.1016/j.intimp.2024.111715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Dendritic cells (DC) play a pivotal role in the onset and progression of immunosenescence-associated diseases, serving as a link between innate and adaptive immunity. Thus, there is a need to establish reference ranges for DC subset levels in healthy adults and investigate the potential impact of age on DC subset levels and phagocytic activity. Single-platform multi-color flow cytometry was performed to assess the proportions of circulating conventional type 1 DC (cDC1), conventional type 2 DC (cDC2), and plasmacytoid DC (pDC), as well as the percentages of CD80, CD86, CD83, PD-L1, and CD32 in cDC1, cDC2, and pDC. Reference ranges were established based on age and gender, and the percentage of circulating DC subsets in different age groups was compared. In addition, circulating DC were enriched using a magnetic bead sorting kit and co-cultured with polystyrene (PS) beads, categorized by age groups, followed by the evaluation of PS bead phagocytosis using light microscopy and flow cytometry. The results indicated that the percentages of circulating cDC1, cDC2, and CD32+cDC2 decreased with age (P < 0.05) and revealed age-related impairment in phagocytic percentage of cDC2 (P < 0.05). These findings provide a deeper understanding of the impact of age on the phenotype and phagocytic activity of DC subsets, shedding light on their role and function in immunosenescence.
Collapse
Affiliation(s)
- Qian Cui
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wentao Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dong Wang
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shuangcui Wang
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Aqing Liu
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guan Zhang
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanjie Yang
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ting Ge
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guixin He
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianchun Yu
- Central Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
4
|
Poznyak AV, Sukhorukov VN, Guo S, Postnov AY, Orekhov AN. Sex Differences Define the Vulnerability to Atherosclerosis. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2023; 17:11795468231189044. [PMID: 37529084 PMCID: PMC10387777 DOI: 10.1177/11795468231189044] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/03/2023] [Indexed: 08/03/2023]
Abstract
For several decades, atherosclerosis has attracted the attention of researchers around the world. Even being a major cause of serious cardiovascular disease and events, atherosclerosis is still not fully understood. Despite the fact that the main players in the pathogenesis of atherosclerosis are well known, many mechanisms of their implementation and interactions remain unknown. The same can be said about the risk factors for atherosclerosis. Many of them are known, but exactly how they work remains to be seen. The main objective of this review is to summarize the latest data on sex as a biological variable in atherosclerosis in humans and animals; to determine what we do not still know about how sex affects the process of growth and complications of atherosclerosis. In this review, we summarized data on sex differences at 3 atherosclerotic aspects: inflammation, vascular remodeling, and plaque morphology. With all overviewed data, we came to the conclusion on the atheroprotective role of female sex.
Collapse
Affiliation(s)
| | - Vasiliy N Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Shuzhen Guo
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese, Beijing, China
| | - Anton Y Postnov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution «Petrovsky National Research Centre of Surgery» (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
5
|
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: 0.7] [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
|
6
|
Inflammatory Cells in Atherosclerosis. Antioxidants (Basel) 2022; 11:antiox11020233. [PMID: 35204116 PMCID: PMC8868126 DOI: 10.3390/antiox11020233] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is a chronic progressive disease that involves damage to the intima, inflammatory cell recruitment and the accumulation of lipids followed by calcification and plaque rupture. Inflammation is considered a key mediator of many events during the development and progression of the disease. Various types of inflammatory cells are reported to be involved in atherosclerosis. In the present paper, we discuss the involved inflammatory cells, their characteristic and functional significance in the development and progression of atherosclerosis. The detailed understanding of the role of all these cells in disease progression at different stages sheds more light on the subject and provides valuable insights as to where and when therapy should be targeted.
Collapse
|
7
|
Depuydt MA, Prange KH, Slenders L, Örd T, Elbersen D, Boltjes A, de Jager SC, Asselbergs FW, de Borst GJ, Aavik E, Lönnberg T, Lutgens E, Glass CK, den Ruijter HM, Kaikkonen MU, Bot I, Slütter B, van der Laan SW, Yla-Herttuala S, Mokry M, Kuiper J, de Winther MP, Pasterkamp G. Microanatomy of the Human Atherosclerotic Plaque by Single-Cell Transcriptomics. Circ Res 2020; 127:1437-1455. [PMID: 32981416 PMCID: PMC7641189 DOI: 10.1161/circresaha.120.316770] [Citation(s) in RCA: 361] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/23/2020] [Accepted: 02/25/2020] [Indexed: 02/01/2023]
Abstract
RATIONALE Atherosclerotic lesions are known for their cellular heterogeneity, yet the molecular complexity within the cells of human plaques has not been fully assessed. OBJECTIVE Using single-cell transcriptomics and chromatin accessibility, we gained a better understanding of the pathophysiology underlying human atherosclerosis. METHODS AND RESULTS We performed single-cell RNA and single-cell ATAC sequencing on human carotid atherosclerotic plaques to define the cells at play and determine their transcriptomic and epigenomic characteristics. We identified 14 distinct cell populations including endothelial cells, smooth muscle cells, mast cells, B cells, myeloid cells, and T cells and identified multiple cellular activation states and suggested cellular interconversions. Within the endothelial cell population, we defined subsets with angiogenic capacity plus clear signs of endothelial to mesenchymal transition. CD4+ and CD8+ T cells showed activation-based subclasses, each with a gradual decline from a cytotoxic to a more quiescent phenotype. Myeloid cells included 2 populations of proinflammatory macrophages showing IL (interleukin) 1B or TNF (tumor necrosis factor) expression as well as a foam cell-like population expressing TREM2 (triggering receptor expressed on myeloid cells 2) and displaying a fibrosis-promoting phenotype. ATACseq data identified specific transcription factors associated with the myeloid subpopulation and T cell cytokine profiles underlying mutual activation between both cell types. Finally, cardiovascular disease susceptibility genes identified using public genome-wide association studies data were particularly enriched in lesional macrophages, endothelial, and smooth muscle cells. CONCLUSIONS This study provides a transcriptome-based cellular landscape of human atherosclerotic plaques and highlights cellular plasticity and intercellular communication at the site of disease. This detailed definition of cell communities at play in atherosclerosis will facilitate cell-based mapping of novel interventional targets with direct functional relevance for the treatment of human disease.
Collapse
Affiliation(s)
- Marie A.C. Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Einsteinweg 55, Leiden, the Netherlands (M.A.C.D., I.B., B.S., J.K.)
| | - Koen H.M. Prange
- Amsterdam University Medical Centers–Location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Meibergdreef 9, the Netherlands (K.H.M.P., M.P.J.d.W.)
| | - Lotte Slenders
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
| | - Tiit Örd
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (T.O., E.A., M.U.K., S.Y.-H.)
| | - Danny Elbersen
- Laboratory for Experimental Cardiology (D.E., S.C.A.d.J), University Medical Center Utrecht, Heidelberglaan 100, the Netherlands
| | - Arjan Boltjes
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
| | - Saskia C.A. de Jager
- Laboratory for Experimental Cardiology (D.E., S.C.A.d.J), University Medical Center Utrecht, Heidelberglaan 100, the Netherlands
| | - Folkert W. Asselbergs
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
| | - Gert J. de Borst
- Vascular Surgery (G.J.d.B.), University Medical Center Utrecht, Heidelberglaan 100, the Netherlands
| | - Einari Aavik
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (T.O., E.A., M.U.K., S.Y.-H.)
| | - Tapio Lönnberg
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland (T.L.)
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Munich, Germany (E.L., M.P.J.d.W.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (E.L., M.P.J.d.W.)
| | - Christopher K. Glass
- Cell and Molecular Medicine (C.K.G.), University of California San Diego, CA
- School of Medicine (C.K.G.), University of California San Diego, CA
| | - Hester M. den Ruijter
- Cardiology (H.M.d.R., M.M.), University Medical Center Utrecht, Heidelberglaan 100, the Netherlands
| | - Minna U. Kaikkonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (T.O., E.A., M.U.K., S.Y.-H.)
| | - Ilze Bot
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Einsteinweg 55, Leiden, the Netherlands (M.A.C.D., I.B., B.S., J.K.)
| | - Bram Slütter
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Einsteinweg 55, Leiden, the Netherlands (M.A.C.D., I.B., B.S., J.K.)
| | - Sander W. van der Laan
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
| | - Seppo Yla-Herttuala
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (T.O., E.A., M.U.K., S.Y.-H.)
| | - Michal Mokry
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
- Cardiology (H.M.d.R., M.M.), University Medical Center Utrecht, Heidelberglaan 100, the Netherlands
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Einsteinweg 55, Leiden, the Netherlands (M.A.C.D., I.B., B.S., J.K.)
| | - Menno P.J. de Winther
- Amsterdam University Medical Centers–Location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Meibergdreef 9, the Netherlands (K.H.M.P., M.P.J.d.W.)
- Institute for Cardiovascular Prevention (IPEK), Munich, Germany (E.L., M.P.J.d.W.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (E.L., M.P.J.d.W.)
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, the Netherlands (L.S., A.B., F.W.A., S.W.v.d.L., M.M., G.P.)
| |
Collapse
|
8
|
Li W, Elshikha AS, Cornaby C, Teng X, Abboud G, Brown J, Zou X, Zeumer-Spataro L, Robusto B, Choi SC, Fredenburg K, Major A, Morel L. T cells expressing the lupus susceptibility allele Pbx1d enhance autoimmunity and atherosclerosis in dyslipidemic mice. JCI Insight 2020; 5:138274. [PMID: 32493841 DOI: 10.1172/jci.insight.138274] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022] Open
Abstract
Patients with systemic lupus erythematosus (SLE) present a high incidence of atherosclerosis, which contributes significantly to morbidity and mortality in this autoimmune disease. An impaired balance between regulatory (Treg) and follicular helper (Tfh) CD4+ T cells is shared by both diseases. However, whether there are common mechanisms of CD4+ T cell dysregulation between SLE and atherosclerosis remains unclear. Pre-B cell leukemia transcription factor 1 isoform d (Pbx1d) is a lupus susceptibility gene that regulates Tfh cell expansion and Treg cell homeostasis. Here, we investigated the role of T cells overexpressing Pbx1d in low-density lipoprotein receptor-deficient (Ldlr-/-) mice fed with a high-fat diet, an experimental model for atherosclerosis. Pbx1d-transgenic T cells exacerbated some phenotypes of atherosclerosis, which were associated with higher autoantibody production, increased Tfh cell frequency, and impaired Treg cell regulation, in Ldlr-/- mice as compared with control T cells. In addition, we showed that dyslipidemia and Pbx1d-transgenic expression independently impaired the differentiation and function of Treg cells in vitro, suggesting a gene/environment additive effect. Thus, our results suggest that the combination of Pbx1d expression in T cells and dyslipidemia exacerbates both atherosclerosis and autoimmunity, at least in part through a dysregulation of Treg cell homeostasis.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Ahmed S Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Sharkia, Egypt
| | - Caleb Cornaby
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Xiangyu Teng
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Josephine Brown
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Xueyang Zou
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA.,Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Leilani Zeumer-Spataro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Brian Robusto
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Seung-Chul Choi
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Kristianna Fredenburg
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Amy Major
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,U.S. Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
9
|
Munjal A, Khandia R. Atherosclerosis: orchestrating cells and biomolecules involved in its activation and inhibition. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 120:85-122. [PMID: 32085889 DOI: 10.1016/bs.apcsb.2019.11.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The term atherosclerosis refers to the condition of deposition of lipids and other substances in and on the artery walls, called as plaque that restricts the normal blood flow. The plaque may be stable or unstable in nature. Unstable plaque can burst and trigger clot formation adding further adversities. The process of plaque formation involves various stages including fatty streak, intermediate or fibro-fatty lesion and advanced lesion. The cells participating in the formation of atherosclerotic plaque include endothelial cells, vascular smooth muscle cells (VSMC), monocytes, monocytes derived macrophages, macrophages and dendritic cells and regulatory T cells (TREG). The role of a variety of cytokines and chemokines have been studied which either help in progression of atherosclerotic plaque or vice versa. The cytokines involved in atherosclerotic plaque formation include IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL-20, IL-25, IL-27, IL-33, IL-37, TNF-α, TGF-β and IFN-γ; whereas amongst the chemokines (family of small cytokines) are CCL2, CCL3, CXCL4, CCL5, CXCL1, CX3CL1, CCL17, CXCL8, CXCL10, CCL20, CCL19 and CCL21 and macrophage migration-inhibitory factor. These are involved in the atherosclerosis advancements, whereas the chemokine CXCL12 is play atheroprotective roles. Apart this, contradictory functions have been documented for few other chemokines such as CXCL16. Since the cytokines and chemokines are amongst the key molecules involved in orchestrating the atherosclerosis advancements, targeting them might be an effective strategy to encumber the atherosclerotic progression. Blockage of cytokines and chemokines via the means of broad-spectrum inhibitors, neutralizing antibodies, usage of decoy receptors or RNA interference have been proved to be useful intervention against atherosclerosis.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| | - Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| |
Collapse
|
10
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
11
|
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
|
12
|
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: 57] [Impact Index Per Article: 8.1] [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
|
13
|
Gil-Pulido J, Zernecke A. Antigen-presenting dendritic cells in atherosclerosis. Eur J Pharmacol 2017; 816:25-31. [DOI: 10.1016/j.ejphar.2017.08.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 11/29/2022]
|
14
|
Wu MY, Li CJ, Hou MF, Chu PY. New Insights into the Role of Inflammation in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2017; 18:2034. [PMID: 28937652 PMCID: PMC5666716 DOI: 10.3390/ijms18102034] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids, smooth muscle cell proliferation, cell apoptosis, necrosis, fibrosis, and local inflammation. Immune and inflammatory responses have significant effects on every phase of atherosclerosis, and increasing evidence shows that immunity plays a more important role in atherosclerosis by tightly regulating its progression. Therefore, understanding the relationship between immune responses and the atherosclerotic microenvironment is extremely important. This article reviews existing knowledge regarding the pathogenesis of immune responses in the atherosclerotic microenvironment, and the immune mechanisms involved in atherosclerosis formation and activation.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Ming-Feng Hou
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Surgery, Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung 807, Taiwan.
- Division of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
15
|
He W, Ren Y, Wang X, Chen Q, Ding S. C reactive protein and enzymatically modified LDL cooperatively promote dendritic cell-mediated T cell activation. Cardiovasc Pathol 2017; 29:1-6. [DOI: 10.1016/j.carpath.2017.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 11/15/2022] Open
|
16
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
17
|
Rai V, Rao VH, Shao Z, Agrawal DK. Dendritic Cells Expressing Triggering Receptor Expressed on Myeloid Cells-1 Correlate with Plaque Stability in Symptomatic and Asymptomatic Patients with Carotid Stenosis. PLoS One 2016; 11:e0154802. [PMID: 27148736 PMCID: PMC4858252 DOI: 10.1371/journal.pone.0154802] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/19/2016] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease with atherosclerotic plaques containing inflammatory cells, including T-lymphocytes, dendritic cells (DCs) and macrophages that are responsible for progression and destabilization of atherosclerotic plaques. Stressed cells undergoing necrosis release molecules that act as endogenous danger signals to alert and activate innate immune cells. In atherosclerotic tissue the number of DCs increases with the progression of the lesion and produce several inflammatory cytokines and growth factors. Triggering receptor expressed on myeloid cells (TREM)-1 plays a crucial role in inflammation. However, relationship of DCs and the role of TREM-1 with the stability of atherosclerotic plaques have not been examined. In this study, we investigated the heterogeneity of the plaque DCs, myeloid (mDC1 and mDC2) and plasmacytoid (pDCs), and examined the expression of TREM-1 and their co-localization with DCs in the plaques from symptomatic (S) and asymptomatic (AS) patients with carotid stenosis. We found increased expression of HLA-DR, fascin, and TREM-1 and decreased expression of TREM-2 and α-smooth muscle actin in S compared to AS atherosclerotic carotid plaques. Both TREM-1 and fascin were co-localized suggesting increased expression of TREM-1 in plaque DCs of S compared to AS patients. These data were supported by increased mRNA transcripts of TREM-1 and decreased mRNA transcripts of TREM-2 in carotid plaques of S compared to AS patients. There was higher density of both CD1c+ mDC1 and CD141+ mDC2 in the carotid plaques from AS compared to S patients, where as the density of CD303+ pDCs were higher in the carotid plaques of S compared to AS patients. These findings suggest a potential role of pDCs and TREM-1 in atherosclerotic plaque vulnerability. Thus, newer therapies could be developed to selectively block TREM-1 for stabilizing atherosclerotic plaques.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Velidi H. Rao
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Zhifei Shao
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
- * E-mail:
| |
Collapse
|
18
|
Chen K, Wang JM, Yuan R, Yi X, Li L, Gong W, Yang T, Li L, Su S. Tissue-resident dendritic cells and diseases involving dendritic cell malfunction. Int Immunopharmacol 2016; 34:1-15. [PMID: 26906720 PMCID: PMC4818737 DOI: 10.1016/j.intimp.2016.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/05/2016] [Indexed: 01/10/2023]
Abstract
Dendritic cells (DCs) control immune responses and are central to the development of immune memory and tolerance. DCs initiate and orchestrate immune responses in a manner that depends on signals they receive from microbes and cellular environment. Although DCs consist mainly of bone marrow-derived and resident populations, a third tissue-derived population resides the spleen and lymph nodes (LNs), different subsets of tissue-derived DCs have been identified in the blood, spleen, lymph nodes, skin, lung, liver, gut and kidney to maintain the tolerance and control immune responses. Tissue-resident DCs express different receptors for microbe-associated molecular patterns (MAMPs) and damage-associated molecular patterns (DAMPs), which were activated to promote the production of pro- or anti-inflammatory cytokines. Malfunction of DCs contributes to diseases such as autoimmunity, allergy, and cancer. It is therefore important to update the knowledge about resident DC subsets and diseases associated with DC malfunction.
Collapse
Affiliation(s)
- Keqiang Chen
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA.
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Ruoxi Yuan
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Xiang Yi
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Liangzhu Li
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Wanghua Gong
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwu Li
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Shaobo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
19
|
Rombouts M, Ammi R, Van Brussel I, Roth L, De Winter BY, Vercauteren SR, Hendriks JMH, Lauwers P, Van Schil PE, De Meyer GRY, Fransen E, Cools N, Schrijvers DM. Linking CD11b (+) Dendritic Cells and Natural Killer T Cells to Plaque Inflammation in Atherosclerosis. Mediators Inflamm 2016; 2016:6467375. [PMID: 27051078 PMCID: PMC4804096 DOI: 10.1155/2016/6467375] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/30/2015] [Accepted: 01/12/2016] [Indexed: 11/25/2022] Open
Abstract
Atherosclerosis remains the leading cause of death and disability in our Western society. To investigate whether the dynamics of leukocyte (sub)populations could be predictive for plaque inflammation during atherosclerosis, we analyzed innate and adaptive immune cell distributions in blood, plaques, and lymphoid tissue reservoirs in apolipoprotein E-deficient (ApoE(-/-)) mice and in blood and plaques from patients undergoing endarterectomy. Firstly, there was predominance of the CD11b(+) conventional dendritic cell (cDC) subset in the plaque. Secondly, a strong inverse correlation was observed between CD11b(+) cDC or natural killer T (NKT) cells in blood and markers of inflammation in the plaque (including CD3, T-bet, CCR5, and CCR7). This indicates that circulating CD11b(+) cDC and NKT cells show great potential to reflect the inflammatory status in the atherosclerotic plaque. Our results suggest that distinct changes in inflammatory cell dynamics may carry biomarker potential reflecting atherosclerotic lesion progression. This not only is crucial for a better understanding of the immunopathogenesis but also bares therapeutic potential, since immune cell-based therapies are emerging as a promising novel strategy in the battle against atherosclerosis and its associated comorbidities. The cDC-NKT cell interaction in atherosclerosis serves as a good candidate for future investigations.
Collapse
Affiliation(s)
- Miche Rombouts
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Rachid Ammi
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Ilse Van Brussel
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Benedicte Y. De Winter
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | | | - Jeroen M. H. Hendriks
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, 2650 Antwerp, Belgium
| | - Patrick Lauwers
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, 2650 Antwerp, Belgium
| | - Paul E. Van Schil
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, 2650 Antwerp, Belgium
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, 2650 Antwerp, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, 2650 Antwerp, Belgium
| | - Dorien M. Schrijvers
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
20
|
Probucol Protects Against Atherosclerosis Through Lipid-lowering and Suppressing Immune Maturation of CD11c+ Dendritic Cells in STZ-induced Diabetic LDLR-/- Mice. J Cardiovasc Pharmacol 2016; 65:620-7. [PMID: 25714599 PMCID: PMC4461394 DOI: 10.1097/fjc.0000000000000234] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Probucol, an agent characterized by lipid-lowering and antioxidant property, retards atherosclerosis effectively. To test the hypothesis that probucol might act its antiatherosclerotic role by suppressing immune maturation of dendritic cells (DCs), 7-week-old LDLR−/− mice were rendered diabetic with streptozotocin (STZ) and then fed either a high-fat diet only or added with 0.5% (wt/wt) probucol for 4 months, and human monocyte-derived dendritic cells were preincubated with or without probucol and stimulated by oxidized low-density lipoprotein. In STZ-induced diabetic LDLR−/− mice, probucol treatment significantly lowered plasma total cholesterol and high-density lipoprotein-cholesterol levels; regressed aortic atherosclerotic lesions; reduced splenic CD40, CD80, CD86, MHC-II expression, and plasma IL-12p70 production; and decreased the expression of CD11c+ DCs within atherosclerotic lesions. In vitro, oxidized low-density lipoprotein promoted human monocyte–derived dendritic cells maturation; stimulated CD40, CD86, CD1a, HLA-DR expression; increased tumor necrosis factor-α production; and decreased IL-4 production. However, these effects were obviously inhibited by probucol pretreatment. In conclusion, our study indicated that probucol effectively retarded atherosclerosis at least partly through lipid-lowering and inhibiting immune maturation of CD11c+ DCs in STZ-induced diabetic LDLR−/− mice.
Collapse
|
21
|
Ciaramella A, Salani F, Bizzoni F, Orfei MD, Caltagirone C, Spalletta G, Bossù P. Myeloid dendritic cells are decreased in peripheral blood of Alzheimer's disease patients in association with disease progression and severity of depressive symptoms. J Neuroinflammation 2016; 13:18. [PMID: 26811068 PMCID: PMC4727381 DOI: 10.1186/s12974-016-0483-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 01/16/2016] [Indexed: 12/22/2022] Open
Abstract
Background Dendritic cells (DCs) are major orchestrators of immune responses and inflammation. They are migratory cells, which may play a role in Alzheimer’s disease (AD), as suggested by prior in vitro studies. With the intent to investigate the clinical relevance of DC modifications in vivo, the present study was aimed to evaluate the levels of blood DCs in AD patients, in relation to the progression of the disease, the severity of its symptoms, and the treatment with acetylcholinesterase inhibitors (AChEIs), a class of drugs used to improve cognitive functioning in people with dementia. Methods The two main subpopulations of immature blood DCs, namely myeloid (mDCs) and plasmacytoid (pDCs) cells, were evaluated by flow cytometry analysis in 106 AD patients, in comparison with the same cells from 65 individuals with mild cognitive impairment (MCI) and 73 healthy control subjects (HC). The relationship between blood DC levels and symptom severity was also assessed in AD patients, and their blood DC frequency was considered both in the absence or presence of treatment with AChEIs. Results A significant depletion in blood mDCs was observed in AD patients, as compared to HC and MCI subjects. At variance, pDC levels were comparable among the three groups of subjects. The mDC decrease was evident only after the emergence of AD clinical symptoms, as confirmed by the follow-up analysis of a subgroup of MCI subjects who exhibited a significant decline in mDCs after their conversion to AD. Notably, the mDC decline was inversely correlated in AD patients with the frequency and severity of depressive symptoms. Eventually, the mDC depletion was not observable in patients treated with AChEIs. Conclusions Our results provide the first evidence that blood mDC levels are dysregulated in AD. This phenomenon appears mainly linked to AD progression, associated with stronger severity of AD-related symptoms, and influenced by AChEI treatment. Taken all together, these data suggest that blood mDCs may serve as a cell source to test disease-induced and treatment-related changes and support the innovative notion that DCs play a role in AD, as ultimate evidence of the immune system participation in disease progression.
Collapse
Affiliation(s)
- Antonio Ciaramella
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy.
| | - Francesca Salani
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy.
| | - Federica Bizzoni
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy.
| | - Maria Donata Orfei
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy.
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy. .,Department of Neuroscience, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Gianfranco Spalletta
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy. .,Neuropsychology Unit, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy. .,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| | - Paola Bossù
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Experimental Neuro-psychobiology Lab, Via Ardeatina 306, 00179, Rome, Italy.
| |
Collapse
|
22
|
Boshuizen MCS, de Winther MPJ. Interferons as Essential Modulators of Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1579-88. [PMID: 25953648 DOI: 10.1161/atvbaha.115.305464] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Abstract
Interferons (IFNs) are key regulators of both innate and adaptive immune responses. The family of IFN cytokines can be divided into 3 main subtypes of which type I and type II IFNs are most well-defined. IFNs are known to be important mediators in atherosclerosis. Evidence from both in vitro and in vivo studies shows that the IFNs are generally proatherosclerotic. However, their role in atherosclerosis is complex, with distinct roles for these cytokines throughout different stages of the disease. In this review, we will discuss the current knowledge on the role of type I and type II IFNs in atherosclerosis development, specifically focusing on their role in endothelial activation, cell recruitment, foam cell formation, and regulation of apoptosis. Furthermore, we will discuss whether IFNs could be considered as new molecular targets for therapeutic intervention in atherosclerosis.
Collapse
Affiliation(s)
- Marieke C S Boshuizen
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Menno P J de Winther
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
23
|
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: 111] [Impact Index Per Article: 11.1] [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
|
24
|
Van Brussel I, Ammi R, Rombouts M, Cools N, Vercauteren SR, De Roover D, Hendriks JMH, Lauwers P, Van Schil PE, Schrijvers DM. Fluorescent activated cell sorting: an effective approach to study dendritic cell subsets in human atherosclerotic plaques. J Immunol Methods 2014; 417:76-85. [PMID: 25527343 DOI: 10.1016/j.jim.2014.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 12/24/2022]
Abstract
Different immune cell types are present within atherosclerotic plaques. Dendritic cells (DC) are of special interest, since they are considered as the 'center of the immuniverse'. Identifying inflammatory DC subtypes within plaques is important for a better understanding of the lesion pathogenesis and pinpoints their contribution to the atherosclerotic process. We have developed a flow cytometry-based method to characterize and isolate different DC subsets (i.e. CD11b(+), Clec9A(+) and CD16(+) conventional (c)DC and CD123(+) plasmacytoid (p)DC) in human atherosclerotic plaques. We revealed a predominance of pro-inflammatory CD11b(+) DC in advanced human lesions, whereas atheroprotective Clec9A(+) DC were almost absent. CD123(+) pDC and CD16(+) DC were also detectable in plaques. Remarkably, plaques from distinct anatomical locations exhibited different cellular compositions: femoral plaques contained less CD11b(+) and Clec9A(+) DC than carotid plaques. Twice as many monocytes/macrophages were observed compared to DC. Moreover, relative amounts of T cells/B cells/NK cells were 6 times as high as DC numbers. For the first time, fluorescent activated cell sorting analysis of DC subsets in human plaques indicated a predominance of CD11b(+) cDC, in comparison with other DC subsets. Isolation of the different subsets will facilitate detailed functional analysis and may have significant implications for tailoring appropriate therapy.
Collapse
Affiliation(s)
- Ilse Van Brussel
- Lab of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Rachid Ammi
- Lab of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Miche Rombouts
- Lab of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Sven R Vercauteren
- Department of Thoracic and Vascular Surgery, ZNA Middelheim (Ziekenhuis Netwerk Antwerpen), Lindendreef 1, B-2020 Antwerp, Belgium
| | - Dominique De Roover
- Department of Thoracic and Vascular Surgery, ZNA Middelheim (Ziekenhuis Netwerk Antwerpen), Lindendreef 1, B-2020 Antwerp, Belgium
| | - Jeroen M H Hendriks
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Edegem, Antwerp, Belgium
| | - Patrick Lauwers
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Edegem, Antwerp, Belgium
| | - Paul E Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Edegem, Antwerp, Belgium
| | - Dorien M Schrijvers
- Lab of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
25
|
Towards non-invasive imaging of vulnerable atherosclerotic plaques by targeting co-stimulatory molecules. Int J Cardiol 2014; 174:503-15. [DOI: 10.1016/j.ijcard.2014.04.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 11/21/2022]
|
26
|
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: 18] [Impact Index Per Article: 1.6] [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
|
27
|
Van Brussel I, Lee WP, Rombouts M, Nuyts AH, Heylen M, De Winter BY, Cools N, Schrijvers DM. Tolerogenic dendritic cell vaccines to treat autoimmune diseases: can the unattainable dream turn into reality? Autoimmun Rev 2014; 13:138-150. [PMID: 24120737 DOI: 10.1016/j.autrev.2013.09.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 09/27/2013] [Indexed: 01/10/2023]
Abstract
Autoimmune diseases affect about one in 15 individuals in developed countries and are characterized by a breakdown in immune tolerance. Current therapeutic approaches against destructive immune responses in autoimmune diseases are based on non-specific agents systemically suppressing the function of many immune effector cells. This indiscriminate immunosuppression, however, often causes serious and sometimes life-threatening side effects. Therefore, the need for more specific treatments resulting in lower toxicity and longer-term solutions is high. Because of the established role of dendritic cells (DCs) in maintaining the balance between immunity and tolerance, tolerogenic (tol)DCs might be novel therapeutic targets to prevent undesirable (auto-)immune responses. The idea behind tolDC therapy is that it is a highly targeted, antigen-specific treatment that only affects the auto-reactive inflammatory response. The therapeutic potential of tolDCs has already been proven in experimental animal models of different autoimmune disorders as well as with in vitro experiments using ex vivo generated human tolDCs, thus the challenge remains in bringing tolDC therapy to the clinic, although first clinical trials have been conducted. In this review, we will extensively discuss the use of tolDCs for induction of antigen-specific tolerance in several autoimmune disease settings, from bench to bedside, including currently applied strategies to generate tolDCs as well as technical difficulties and challenges in the field.
Collapse
Affiliation(s)
- Ilse Van Brussel
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
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
|
29
|
Dietel B, Cicha I, Achenbach S, Kollmar R, Garlichs C, Tauchi M. Different treatment settings of Granulocyte-Colony Stimulating Factor and their impact on T cell-specific immune response in experimental stroke. Immunol Lett 2013; 158:95-100. [PMID: 24333341 DOI: 10.1016/j.imlet.2013.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Cerebral ischemia is associated with infectious complications due to immunosuppression and decreased T lymphocyte activity. G-CSF, which has neuroprotective properties, is known to modulate inflammatory processes after induced stroke. The aim of our study was to investigate the impact of G-CSF in experimental stroke and to compare two different modes of treatment, focusing on circulating T lymphocytes. METHODS Cerebral ischemia was induced in Wistar rats by occlusion of the middle cerebral artery, followed by reperfusion after 1h. G-CSF was applied either as a single dose 30 min after occlusion, or daily for seven days. Silver staining was used to determine infarct size. T lymphocytes in the peripheral blood were measured before and 7 days after induced cerebral ischemia by flow cytometry. In addition, migration of CD3-expressing T lymphocytes into the brain was investigated by immunohistochemistry. RESULTS Both single dose and daily treatment with G-CSF significantly reduced infarct size. A significant improvement of neurological outcome was only observed after single application of G-CSF. While a decrease in peripheral T lymphocytes was detected seven days after induced stroke, no reduction was observed in the G-CSF-treated groups. Apart from that, G-CSF significantly reduced the number of brain migrated T lymphocytes in both treatment settings as compared to vehicle. CONCLUSION A single dose of G-CSF exerted neuroprotective effects in ischemic stroke, which were less pronounced after daily G-CSF application. Both treatment strategies inhibited stroke-induced reduction of T lymphocytes in peripheral blood, which may have contributed to the reduction of infarct size.
Collapse
Affiliation(s)
- Barbara Dietel
- Department of Cardiology and Angiology, University Hospital Erlangen-Nuremberg, Erlangen, Germany.
| | - Iwona Cicha
- Department of Cardiology and Angiology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Stephan Achenbach
- Department of Cardiology and Angiology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Rainer Kollmar
- Department of Neurology, University Hospital Erlangen-Nuremberg, Erlangen, Germany; Department of Neurology, Hospital Darmstadt, Darmstadt, Germany
| | - Christoph Garlichs
- Department of Cardiology and Angiology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| | - Miyuki Tauchi
- Department of Neurology, University Hospital Erlangen-Nuremberg, Erlangen, Germany; Division of Molecular Neurology, University Hospital Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
30
|
Alberts-Grill N, Denning TL, Rezvan A, Jo H. The role of the vascular dendritic cell network in atherosclerosis. Am J Physiol Cell Physiol 2013; 305:C1-21. [PMID: 23552284 DOI: 10.1152/ajpcell.00017.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A complex role has been described for dendritic cells (DCs) in the potentiation and control of vascular inflammation and atherosclerosis. Resident vascular DCs are found in the intima of atherosclerosis-prone vascular regions exposed to disturbed blood flow patterns. Several phenotypically and functionally distinct vascular DC subsets have been described. The functional heterogeneity of these cells and their contributions to vascular homeostasis, inflammation, and atherosclerosis are only recently beginning to emerge. Here, we review the available literature, characterizing the origin and function of known vascular DC subsets and their important role contributing to the balance of immune activation and immune tolerance governing vascular homeostasis under healthy conditions. We then discuss how homeostatic DC functions are disrupted during atherogenesis, leading to atherosclerosis. The effectiveness of DC-based "atherosclerosis vaccine" therapies in the treatment of atherosclerosis is also reviewed. We further provide suggestions for distinguishing DCs from macrophages and discuss important future directions for the field.
Collapse
Affiliation(s)
- Noah Alberts-Grill
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
31
|
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
|
32
|
Rosenfeld ME. Inflammation and atherosclerosis: direct versus indirect mechanisms. Curr Opin Pharmacol 2013; 13:154-60. [PMID: 23357128 DOI: 10.1016/j.coph.2013.01.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/05/2013] [Accepted: 01/07/2013] [Indexed: 12/15/2022]
Abstract
It is now widely accepted that the development of atherosclerotic lesions involves a chronic inflammatory response that includes both innate and adaptive immune mechanisms. However, it is still unclear precisely what induces the inflammatory response. Furthermore, inflammation within the blood vessel can be divided into direct mechanisms where the primary inflammatory events occur within the intima of the blood vessel and contribute to both the initiation and progression of the plaques and indirect mechanisms where inflammation at nonvascular sites can contribute to the progression of the lesions. The direct mechanisms include lipid deposition and modification, influx of lipoprotein associated factors and microparticles derived from many different cell types, and possibly bacterial and viral infection of vascular cells. Indirect mechanisms derive from inflammation related to autoimmune diseases, smoking, respiratory infection, and pollution exposure, and possibly periodontal disease and gastric infection. The mechanisms include secretion of cytokines and other inflammatory factors into the circulation with subsequent uptake into the plaques, egress and recruitment of activated inflammatory cells, formation of dysfunctional HDL and crossreactive autoantibodies.
Collapse
|
33
|
Yang Q, Chen Y, Yong W. FOXP3 genetic variant and risk of acute coronary syndrome in Chinese Han population. Cell Biochem Funct 2013; 31:599-602. [PMID: 23299803 DOI: 10.1002/cbf.2945] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 11/15/2012] [Accepted: 11/28/2012] [Indexed: 02/05/2023]
Abstract
Coronary artery disease is the most common type of heart disease and a leading cause of morbidity and mortality all over the world. Acute coronary syndrome (ACS) is the most serious form of coronary artery disease. Recently, many studies indicated that genetic susceptibility may play a vital role in the pathogenesis of coronary heart disease including ACS. Forkhead/winged helix transcription factor (FOXP3) gene polymorphisms have been previously found to be associated with inflammatory diseases. To determine whether FOXP3 polymorphisms are associated with ACS, we examined the single nucleotide polymorphism rs3761548 of FOXP3 gene by polymerase chain reaction-polyacrylamide gel electrophoresis in 226 ACS patients and 259 unrelated healthy subjects. Our results showed that single nucleotide polymorphism rs3761548 had association with ACS in Chinese Han population. These data indicate that, for the first time, FOXP3 gene polymorphism may appear to play an important role in the susceptibility of ACS in Chinese Han population.
Collapse
Affiliation(s)
- Qing Yang
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | | | | |
Collapse
|
34
|
Translocator protein (18 kDa): a promising therapeutic target and diagnostic tool for cardiovascular diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:162934. [PMID: 23251719 PMCID: PMC3516045 DOI: 10.1155/2012/162934] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/22/2012] [Accepted: 11/04/2012] [Indexed: 01/15/2023]
Abstract
The translocator protein (18 kDa) (TSPO) is a five transmembrane domain protein in mitochondria, abundantly expressed in a variety of organs and tissues. TSPO contributes to a wide range of biological processes, including cholesterol transportation, mitochondrial membrane potential and respiratory chain regulation, apoptosis, and oxidative stress. Recent studies have demonstrated that TSPO might also be involved in the physiological regulation of cardiac chronotropy and inotropy. Accordingly, TSPO ligands play significant roles in protecting the cardiovascular systems under pathological conditions through cardiac electrical activity retention, intracellular calcium maintenance, mitochondrial energy provision, mitochondrial membrane potential equilibrium, and reactive oxygen species inhibition. This paper focuses on the physiological and pathological characteristics of TSPO in the cardiovascular systems and also summarizes the properties of TSPO ligands. TSPO represents a potential therapeutic target and diagnostic tool for cardiovascular diseases including arrhythmia, myocardial infarction, cardiac hypertrophy, atherosclerosis, myocarditis, and large vessel vasculitis.
Collapse
|
35
|
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: 0.5] [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
|
36
|
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: 0.8] [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
|
37
|
Transcript and protein analysis reveals better survival skills of monocyte-derived dendritic cells compared to monocytes during oxidative stress. PLoS One 2012; 7:e43357. [PMID: 22916248 PMCID: PMC3419731 DOI: 10.1371/journal.pone.0043357] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 07/23/2012] [Indexed: 11/19/2022] Open
Abstract
Background Dendritic cells (DCs), professional antigen-presenting cells with the unique ability to initiate primary T-cell responses, are present in atherosclerotic lesions where they are exposed to oxidative stress that generates cytotoxic reactive oxygen species (ROS). A large body of evidence indicates that cell death is a major modulating factor of atherogenesis. We examined antioxidant defence systems of human monocyte-derived (mo)DCs and monocytes in response to oxidative stress. Methods Oxidative stress was induced by addition of tertiary-butylhydroperoxide (tert-BHP, 30 min). Cellular responses were evaluated using flow cytometry and confocal live cell imaging (both using 5-(and-6)-chloromethyl-2,7-dichlorodihydrofluorescein diacetate, CM-H2DCFDA). Viability was assessed by the neutral red assay. Total RNA was extracted for a PCR profiler array. Five genes were selected for confirmation by Taqman gene expression assays, and by immunoblotting or immunohistochemistry for protein levels. Results Tert-BHP increased CM-H2DCFDA fluorescence and caused cell death. Interestingly, all processes occurred more slowly in moDCs than in monocytes. The mRNA profiler array showed more than 2-fold differential expression of 32 oxidative stress–related genes in unstimulated moDCs, including peroxiredoxin-2 (PRDX2), an enzyme reducing hydrogen peroxide and lipid peroxides. PRDX2 upregulation was confirmed by Taqman assays, immunoblotting and immunohistochemistry. Silencing PRDX2 in moDCs by means of siRNA significantly increased CM-DCF fluorescence and cell death upon tert-BHP-stimulation. Conclusions Our results indicate that moDCs exhibit higher intracellular antioxidant capacities, making them better equipped to resist oxidative stress than monocytes. Upregulation of PRDX2 is involved in the neutralization of ROS in moDCs. Taken together, this points to better survival skills of DCs in oxidative stress environments, such as atherosclerotic plaques.
Collapse
|
38
|
Role of Peroxisome Proliferator-Activated Receptor-γ in Vascular Inflammation. Int J Vasc Med 2012; 2012:508416. [PMID: 22888436 PMCID: PMC3409528 DOI: 10.1155/2012/508416] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 06/08/2012] [Indexed: 12/13/2022] Open
Abstract
Vascular inflammation plays a crucial role in atherosclerosis, and its regulation is important to prevent cerebrovascular and coronary artery disease. The inflammatory process in atherogenesis involves a variety of immune cells including monocytes/macrophages, lymphocytes, dendritic cells, and neutrophils, which all express peroxisome proliferator-activated receptor-γ (PPAR-γ). PPAR-γ is a nuclear receptor and transcription factor in the steroid superfamily and is known to be a key regulator of adipocyte differentiation. Increasing evidence from mainly experimental studies has demonstrated that PPAR-γ activation by endogenous and synthetic ligands is involved in lipid metabolism and anti-inflammatory activity. In addition, recent clinical studies have shown a beneficial effect of thiazolidinediones, synthetic PPAR-γ ligands, on cardiovascular disease beyond glycemic control. These results suggest that PPAR-γ activation is an important regulator in vascular inflammation and is expected to be a therapeutic target in the treatment of atherosclerotic complications. This paper reviews the recent findings of PPAR-γ involvement in vascular inflammation and the therapeutic potential of regulating the immune system in atherosclerosis.
Collapse
|
39
|
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: 1.0] [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
|
40
|
Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm 2012; 2012:690643. [PMID: 22851815 PMCID: PMC3407661 DOI: 10.1155/2012/690643] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/17/2012] [Indexed: 02/08/2023] Open
Abstract
Earlier investigations have revealed a surprising complexity and variety in the range of interaction between cells of the innate and adaptive immune system. Our understanding of the specialized roles of dendritic cell (DC) subsets in innate and adaptive immune responses has been significantly advanced over the years. Because of their immunoregulatory capacities and because very small numbers of activated DC are highly efficient at generating immune responses against antigens, DCs have been vigorously used in clinical trials in order to elicit or amplify immune responses against cancer and chronic infectious diseases. A better insight in DC immunobiology and function has stimulated many new ideas regarding the potential ways forward to improve DC therapy in a more fundamental way. Here, we discuss the continuous search for optimal in vitro conditions in order to generate clinical-grade DC with a potent immunogenic potential. For this, we explore the molecular and cellular mechanisms underlying adequate immune responses and focus on most favourable DC culture regimens and activation stimuli in humans. We envisage that by combining each of the features outlined in the current paper into a unified strategy, DC-based vaccines may advance to a higher level of effectiveness.
Collapse
|
41
|
Humoral and cellular immune responses in atherosclerosis: Spotlight on B- and T-cells. Vascul Pharmacol 2012; 56:193-203. [DOI: 10.1016/j.vph.2012.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/17/2012] [Accepted: 01/28/2012] [Indexed: 01/20/2023]
|