1
|
Reilly NA, Dekkers KF, Molenaar J, Arumugam S, Kuipers TB, Ariyurek Y, Hoeksema MA, Jukema JW, Heijmans BT. EPA Induces an Anti-Inflammatory Transcriptome in T Cells, Implicating a Triglyceride-Independent Pathway in Cardiovascular Risk Reduction. JACC Basic Transl Sci 2025; 10:383-395. [PMID: 40139879 PMCID: PMC12013851 DOI: 10.1016/j.jacbts.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 03/29/2025]
Abstract
Twice-daily intake of purified eicosapentaenoic acid (EPA) reduces atherosclerotic cardiovascular disease risk in patients with high triglycerides, but its exact mechanism remains unclear. We exposed non-activated CD4+ T cells to 100μM EPA, oleic acid, palmitic acid, or control, and conducted RNA and ATAC-sequencing after 48 hours. EPA exposure downregulated immune response-related genes like HLA-DRA, CD69, and IL2RA, and upregulated oxidative stress prevention genes like NQO1. Transcription factor footprinting showed decreased GATA3 and PU.1, and increased REV-ERB. These effects were specific to EPA, suggesting it induces an anti-inflammatory transcriptomic landscape in CD4+ T cells, contributing to its observed cardiovascular benefits.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen Molenaar
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sinthuja Arumugam
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas B Kuipers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Yavuz Ariyurek
- Leiden Genome Technology Center, Department of Human Genetics, Leiden University Medical Center, the Netherlands
| | - Marten A Hoeksema
- Department of Medical Biochemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Picone F, Giudice V, Iside C, Venturini E, Di Pietro P, Vecchione C, Selleri C, Carrizzo A. Lymphocyte Subset Imbalance in Cardiometabolic Diseases: Are T Cells the Missing Link? Int J Mol Sci 2025; 26:868. [PMID: 39940640 PMCID: PMC11816853 DOI: 10.3390/ijms26030868] [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: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiometabolic and cardiovascular diseases (CVDs) remain the leading cause of death worldwide, with well-established risk factors such as smoking, obesity, and diabetes contributing to plaque formation and chronic inflammation. However, emerging evidence suggests that the immune system plays a more significant role in the development and progression of CVD than previously thought. Specifically, the finely tuned regulation of lymphocyte subsets governs post-injury inflammation and tissue damage resolution and orchestrates the functions and activation of endothelial cells, cardiomyocytes, and fibroblasts in CVD-associated lesions (e.g., atherosclerotic plaques). A deeper understanding of the immune system's involvement in CVD development and progression will provide new insights into disease biology and uncover novel therapeutic targets aimed at re-establishing immune homeostasis. In this review, we summarize the current state of knowledge on the distribution and involvement of lymphocyte subsets in CVD, including atherosclerosis, diabetes, hypertension, myocardial infarction, and stroke.
Collapse
Affiliation(s)
- Francesca Picone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Concetta Iside
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | | | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| |
Collapse
|
3
|
Merdler I, Chitturi KR, Chaturvedi A, Lupu L, Wallace R, Cellamare M, Zhang C, Sawant V, Ben-Dor I, Case BC, Waksman R. Examining the relationship between monocytes and monocyte-derived ratios in post-percutaneous coronary intervention patients and their impact on coronary artery disease progression. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2025:S1553-8389(25)00023-5. [PMID: 39894723 DOI: 10.1016/j.carrev.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Inflammation plays a key role in the progression and instability of coronary atherosclerosis. Monocytes and their ratios with eosinophils and lymphocytes serve as valuable markers for assessing inflammation. We explored blood monocyte levels and their related ratios in patients undergoing percutaneous coronary intervention (PCI) for in-stent restenosis (ISR) or significant de novo lesions (DNL). METHODS AND RESULTS A total of 3912 PCI procedures were identified from a single-center retrospective registry (2013-2022) and categorized into three groups: single PCI and no subsequent intervention (control group, n = 3342), significant ISR requiring repeat PCI (ISR-PCI group, n = 219), and significant de novo lesions requiring repeat PCI (DNL-PCI group, n = 351). Monocyte counts and monocyte-related ratios were evaluated at the index procedure and follow-up (clinical or repeat PCI procedures). Comorbidities were more prevalent in the ISR-PCI and DNL-PCI groups than those in the control group. In comparison to the control group, both ISR-PCI (15.6 ± 26.7 vs. 24.4 ± 37.8, P < 0.001) and DNL-PCI groups (16.2 ± 28.5 vs. 24.4 ± 37.8, P < 0.001) exhibited a significantly lower baseline monocyte-to-eosinophil ratio. In the adjusted regression models, a lower baseline monocyte-to-eosinophil ratio (P = 0.001) and monocyte-to-lymphocyte ratio (P = 0.04) were associated with DNL, whereas no such association was observed in ISR-PCI cases (P = 0.4 for both ratios). CONCLUSION Our findings reinforce the role of inflammatory markers, such as monocytes and monocyte-related ratios, in identifying individuals at risk for the progression of coronary disease post-PCI.
Collapse
Affiliation(s)
- Ilan Merdler
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Kalyan R Chitturi
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Abhishek Chaturvedi
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Lior Lupu
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Ryan Wallace
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Matteo Cellamare
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Cheng Zhang
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Vaishnavi Sawant
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Itsik Ben-Dor
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Brian C Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Ron Waksman
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
4
|
Lu Y, Wu S, Zhu S, Shen J, Liu C, Zhao C, Su S, Ma H, Xiang M, Xie Y. Integrated Single-Cell Analysis Revealed Novel Subpopulations of Foamy Macrophages in Human Atherosclerotic Plaques. Biomolecules 2024; 14:1606. [PMID: 39766313 PMCID: PMC11675068 DOI: 10.3390/biom14121606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Foam cell formation is a hallmark of atherosclerosis, yet the cellular complexity within foam cells in human plaques remains unexplored. Here, we integrate published single-cell RNA-sequencing, spatial transcriptomic, and chromatin accessibility sequencing datasets of human atherosclerotic lesions across eight distinct studies. Through this large-scale integration of patient-derived information, we identified foamy macrophages enriched for genes characteristic of the foamy signature. We further re-clustered the foamy macrophages into five unique subsets with distinct potential functions: (i) pro-foamy macrophages, exhibiting relatively high inflammatory and adhesive properties; (ii) phagocytic foamy macrophages, specialized in efferocytosis; (iii) high-efflux foamy macrophages marked by high NR1H3 expression; (iv) mature foamy macrophages prone to programmed cell death; and (v) synthetic subset. Trajectory analysis elucidated a bifurcated differentiation cell fate from pro-foam macrophages toward either the programmed death (iv) or synthetic (v) phenotype. The existence of these foamy macrophage subsets was validated by immunostaining. Moreover, these foamy macrophage subsets exhibited strong potential ligand-receptor interactions. Finally, we conducted Mendelian randomization analyses to identify a possible causal relationship between key regulatory genes along the programmed death pathway in foamy macrophages and atherosclerotic diseases. This study provides a high-resolution map of foam cell diversity and a set of potential key regulatory genes in atherosclerotic plaques, offering novel insights into the multifaceted pathophysiology underlying human atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Meixiang Xiang
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| | - Yao Xie
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| |
Collapse
|
5
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [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: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
6
|
Zhang W, Cai Z, Ma D, Liu M, Wang J, Sun L, Lew AM, Xu Y. Local adaptive immunity in atherosclerosis with T cell activation by aortic dendritic cells accelerates pathogenesis. iScience 2024; 27:111144. [PMID: 39502289 PMCID: PMC11536043 DOI: 10.1016/j.isci.2024.111144] [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: 04/04/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Atherosclerosis represents a chronic inflammatory condition in arterial walls, where local immune cells significantly contribute to disease progression. This study employed various in situ immunological techniques to investigate the specific roles of aortic dendritic cell (DC) subsets in atherosclerotic animal models, distinguishing between normal and diseased immune contexts. Our findings revealed that aortic DCs, particularly the cDC1 subset, played a critical role in facilitating CD8+ T cell activation through antigen presentation. Additionally, atherosclerosis-induced increases in GM-CSF levels enhanced CCR7 expression on aortic monocyte-derived DCs, promoting their recruitment and IL-12 production for Th1 differentiation. Notably, immunizing pre-atherosclerotic mice with DC-presented antigens or transferring aortic DCs from atherosclerotic mice resulted in accelerated disease onset. This research elucidates the adaptive immune functions of aortic DCs, offering insights into the cellular mechanisms driving aortic inflammation and potential therapeutic targets for atherosclerosis management.
Collapse
Affiliation(s)
- Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Zecheng Cai
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Dan Ma
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Meng Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Andrew M. Lew
- The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville, VIC, Australia
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| |
Collapse
|
7
|
Ahmed ME, Leistner DM, Hakim D, Abdelwahed Y, Coskun AU, Maynard C, Seppelt C, Nelles G, Meteva D, Cefalo NV, Libby P, Landmesser U, Stone PH. Endothelial Shear Stress Metrics Associate With Proinflammatory Pathways at the Culprit Site of Coronary Erosion. JACC Basic Transl Sci 2024; 9:1269-1283. [PMID: 39619137 PMCID: PMC11604495 DOI: 10.1016/j.jacbts.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 01/16/2025]
Abstract
Low endothelial shear stress (ESS) and associated adverse biomechanical features stimulate inflammation, contribute to atherogenesis, and predispose to coronary plaque disruption. The mechanistic links between adverse flow-related hemodynamics and inflammatory mediators implicated in plaque erosion, however, remain little explored. We investigated the relationship of high-risk ESS metrics to culprit lesion proinflammatory/proatherogenic cells and cytokines/chemokines implicated in coronary plaque erosion in patients with acute coronary syndromes. In eroded plaques, low ESS, high ESS gradient, and steepness of plaque topographical slope associated with increased numbers of local T cells and subsets (CD4+, CD8+, natural killer T cells) as well as inflammatory mediators (interleukin [IL]-6, macrophage inflammatory protein-1β, IL-1β, IL-2).
Collapse
Affiliation(s)
- Mona E. Ahmed
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Heart and Vascular Center, Karolinska University Hospital, Stockholm, Sweden
| | - David M. Leistner
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Rhine Main, Frankfurt, Germany
- Berlin Institute of Health, Berlin, Germany
- Department of Cardiology and Angiology, Goethe University, Frankfurt am Main, Germany
| | - Diaa Hakim
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Youssef Abdelwahed
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin, Germany
| | | | | | - Claudio Seppelt
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Rhine Main, Frankfurt, Germany
- Department of Cardiology and Angiology, Goethe University, Frankfurt am Main, Germany
| | - Gregor Nelles
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Rhine Main, Frankfurt, Germany
| | - Denitsa Meteva
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin, Germany
| | - Nicholas V. Cefalo
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ulf Landmesser
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Rhine Main, Frankfurt, Germany
| | - Peter H. Stone
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Hmiel L, Zhang S, Obare LM, Santana MADO, Wanjalla CN, Titanji BK, Hileman CO, Bagchi S. Inflammatory and Immune Mechanisms for Atherosclerotic Cardiovascular Disease in HIV. Int J Mol Sci 2024; 25:7266. [PMID: 39000373 PMCID: PMC11242562 DOI: 10.3390/ijms25137266] [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: 06/01/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Atherosclerotic vascular disease disproportionately affects persons living with HIV (PLWH) compared to those without. The reasons for the excess risk include dysregulated immune response and inflammation related to HIV infection itself, comorbid conditions, and co-infections. Here, we review an updated understanding of immune and inflammatory pathways underlying atherosclerosis in PLWH, including effects of viral products, soluble mediators and chemokines, innate and adaptive immune cells, and important co-infections. We also present potential therapeutic targets which may reduce cardiovascular risk in PLWH.
Collapse
Affiliation(s)
- Laura Hmiel
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Suyu Zhang
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Boghuma K. Titanji
- Division of Infectious Diseases, Emory University, Atlanta, GA 30322, USA
| | - Corrilynn O. Hileman
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Shashwatee Bagchi
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
Reilly NA, Sonnet F, Dekkers KF, Kwekkeboom JC, Sinke L, Hilt S, Suleiman HM, Hoeksema MA, Mei H, van Zwet EW, Everts B, Ioan-Facsinay A, Jukema JW, Heijmans BT. Oleic acid triggers metabolic rewiring of T cells poising them for T helper 9 differentiation. iScience 2024; 27:109496. [PMID: 38558932 PMCID: PMC10981094 DOI: 10.1016/j.isci.2024.109496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/29/2023] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are the most common immune cells in atherosclerotic plaques, and the function of T cells can be altered by fatty acids. Here, we show that pre-exposure of CD4+ T cells to oleic acid, an abundant fatty acid linked to cardiovascular events, upregulates core metabolic pathways and promotes differentiation into interleukin-9 (IL-9)-producing cells upon activation. RNA sequencing of non-activated T cells reveals that oleic acid upregulates genes encoding key enzymes responsible for cholesterol and fatty acid biosynthesis. Transcription footprint analysis links these expression changes to the differentiation toward TH9 cells, a pro-atherogenic subset. Spectral flow cytometry shows that pre-exposure to oleic acid results in a skew toward IL-9+-producing T cells upon activation. Importantly, pharmacological inhibition of either cholesterol or fatty acid biosynthesis abolishes this effect, suggesting a beneficial role for statins beyond cholesterol lowering. Taken together, oleic acid may affect inflammatory diseases like atherosclerosis by rewiring T cell metabolism.
Collapse
Affiliation(s)
- Nathalie A. Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Friederike Sonnet
- Leiden University Center for Infectious Diseases (LUCID), Leiden, the Netherlands
| | - Koen F. Dekkers
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | | | - Lucy Sinke
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Stan Hilt
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Hayat M. Suleiman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Marten A. Hoeksema
- Department of Medical Biochemistry, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Erik W. van Zwet
- Medical Statistics, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Bart Everts
- Leiden University Center for Infectious Diseases (LUCID), Leiden, the Netherlands
| | - Andreea Ioan-Facsinay
- Department of Rheumatology Leiden University Medical Center, Leiden, the Netherlands
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bastiaan T. Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, the Netherlands
| |
Collapse
|
10
|
Miyazaki S, Fujisue K, Yamanaga K, Sueta D, Usuku H, Tabata N, Ishii M, Hanatani S, Hoshiyama T, Kanazawa H, Takashio S, Arima Y, Araki S, Yamamoto E, Matsushita K, Tsujita K. Prognostic Significance of Soluble PD-L1 on Cardiovascular Outcomes in Patients with Coronary Artery Disease. J Atheroscler Thromb 2024; 31:355-367. [PMID: 37793811 PMCID: PMC10999719 DOI: 10.5551/jat.64183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 08/03/2023] [Indexed: 10/06/2023] Open
Abstract
AIMS Programmed cell death-1 (PD-1) and its ligand (PD-L1) regulate T cells, leading to immunotolerance. We previously demonstrated that patients with coronary artery disease (CAD) had increased circulating levels of soluble PD-L1 (sPD-L1). However, the prognostic significance of sPD-L1 on cardiovascular outcomes is unknown. In the present study, we evaluated the association between sPD-L1 and cardiovascular events in patients with CAD. METHODS We prospectively measured sPD-L1 in patients with CAD admitted to Kumamoto University Hospital between December 2017 and January 2020 and observed their cardiovascular event rate. The primary outcome was a composite of death from non-cardiovascular causes, death from cardiovascular causes, non-fatal myocardial infarction, unstable angina pectoris, revascularization, hospitalization for heart failure, and ischemic stroke. RESULTS Finally, 627 patients were enrolled, and 35 patients were lost to follow-up. The median follow-up duration was 522 days. In total, 124 events were recorded. The Kaplan-Meier curve showed that the event rate was higher in the higher sPD-L1 group (median ≥ 136 pg/dL) than in the lower sPD-L1 group (25.0% vs. 16.9%; p=0.028, log-rank test). Univariate Cox proportional hazards analysis showed that high-sensitivity C-reactive protein, an estimated glomerular filtration rate of <60 mL/min/1.73m2, B-type natriuretic peptide, left ventricular ejection fraction, and sPD-L1 were significantly associated with cardiovascular events. Multivariable Cox proportional hazards analysis of factors that were significant in univariate analysis identified that sPD-L1 was significantly and independently associated with cardiovascular events (hazard ratio: 1.364, 95% confidence interval: 1.018-1.828, p=0.038). CONCLUSIONS Higher sPD-L1 levels were significantly associated with future cardiovascular events in patients with CAD.
Collapse
Affiliation(s)
- Shuhei Miyazaki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Kenshi Yamanaga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| | - Kenichi Matsushita
- Division of Advanced Cardiovascular Therapeutics, Department of Cardiovascular Medicine, Kumamoto University Hospital,
Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences and Center for Metabolic Regulation of Healthy Aging (CMHA), Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
12
|
Winchester NE, Panigrahi S, Haria A, Chakraborty A, Su X, Chen B, Morris SR, Clagett BM, Juchnowski SM, Yadavalli R, Villinger F, Paiardini M, Harth K, Kashyap VS, Calabrese LH, Margolis L, Sieg SF, Shive CL, Gianella S, Funderburg NT, Zidar DA, Lederman MM, Freeman ML. Cytomegalovirus Infection Facilitates the Costimulation of CD57+CD28- CD8 T Cells in HIV Infection and Atherosclerosis via the CD2-LFA-3 Axis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:245-257. [PMID: 38047900 PMCID: PMC10843654 DOI: 10.4049/jimmunol.2300267] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023]
Abstract
CD8 T cells are emerging as important mediators in atherosclerosis and cardiovascular disease (CVD). Immune activation may play a particular role in people with HIV (PWH) who are at an increased risk of CVD, even after controlling for known CVD risk factors. Latent CMV infection is associated with increased CVD risk for both PWH and people without HIV, and human CMV-specific CD4 and CD8 T cells are enriched for an immunosenescent phenotype. We previously showed that CMV coinfection in PWH promotes vascular homing and activation of inflammatory CD4 T cells through the CD2-LFA-3 axis. However, the role of CD2/LFA3 costimulation of CD8 T cells in PWH with CMV has yet to be described. In the present study, we demonstrate that CD2 expression on CX3CR1+CD57+CD28- inflammescent CD8 T cells is increased on cells from CMV-seropositive PWH. In vitro CD2/LFA-3 costimulation enhances TCR-mediated activation of these inflammatory CD8 memory T cells. Finally, we show that LFA-3 is highly expressed in aortas of SIV-infected rhesus macaques and in atherosclerotic plaques of people without HIV. Our findings are consistent with a model in which CMV infection enhances CD2 expression on highly proinflammatory CD8 T cells that can then be stimulated by LFA-3 expressed in the vasculature, even in the absence of CD28 costimulation. This model, in which CMV infection exacerbates toxic cytokine and granzyme production by CD8 T cells within the vasculature, highlights a potential therapeutic target in atherosclerosis development and progression, especially for PWH.
Collapse
Affiliation(s)
- Nicole E. Winchester
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - Soumya Panigrahi
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Anokhi Haria
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Archeesha Chakraborty
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Xi Su
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Bonnie Chen
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Stephen R. Morris
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Brian M. Clagett
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Steven M. Juchnowski
- Division of Cardiology, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Raghavendra Yadavalli
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Karem Harth
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - Vikram S. Kashyap
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - Leonard H. Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Leonid Margolis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Scott F. Sieg
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Carey L. Shive
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas T. Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, USA
| | - David A. Zidar
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - Michael M. Lederman
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Michael L. Freeman
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
13
|
Potashnikova D, Maryukhnich E, Vorobyeva D, Rusakovich G, Komissarov A, Tvorogova A, Gontarenko V, Vasilieva E. Cytokine Profiling of Plasma and Atherosclerotic Plaques in Patients Undergoing Carotid Endarterectomy. Int J Mol Sci 2024; 25:1030. [PMID: 38256102 PMCID: PMC10816498 DOI: 10.3390/ijms25021030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Atherosclerotic plaques are sites of chronic inflammation with diverse cell contents and complex immune signaling. Plaque progression and destabilization are driven by the infiltration of immune cells and the cytokines that mediate their interactions. Here, we attempted to compare the systemic cytokine profiles in the blood plasma of patients with atherosclerosis and the local cytokine production, using ex vivo plaque explants from the same patients. The developed method of 41-plex xMAP data normalization allowed us to differentiate twenty-two cytokines produced by the plaque that were not readily detectable in free circulation and six cytokines elevated in blood plasma that may have other sources than atherosclerotic plaque. To verify the xMAP data on the putative atherogenesis-driving chemokines MCP-1 (CCL2), MIP-1α (CCL3), MIP-1β (CCL4), RANTES (CCL5), and fractalkine (CX3CL1), qPCR was performed. The MIP1A (CCL3), MIP1B (CCL4), FKN (CX3CL1), and MCP1 (CCL2) genes were expressed at high levels in the plaques, whereas RANTES (CCL5) was almost absent. The expression patterns of the chemokines were restricted to the plaque cell types: the MCP1 (CCL2) gene was predominantly expressed in endothelial cells and monocytes/macrophages, MIP1A (CCL3) in monocytes/macrophages, and MIP1B (CCL4) in monocytes/macrophages and T cells. RANTES (CCL5) was restricted to T cells, while FKN (CX3CL1) was not differentially expressed. Taken together, our data indicate a plaque-specific cytokine production profile that may be a useful tool in atherosclerosis studies.
Collapse
Affiliation(s)
- Daria Potashnikova
- Laboratory of Atherothrombosis, Cardiology Department, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127006 Moscow, Russia
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| | - Elena Maryukhnich
- Laboratory of Atherothrombosis, Cardiology Department, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127006 Moscow, Russia
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| | - Daria Vorobyeva
- Laboratory of Atherothrombosis, Cardiology Department, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127006 Moscow, Russia
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| | - George Rusakovich
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| | - Alexey Komissarov
- Laboratory of Atherothrombosis, Cardiology Department, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127006 Moscow, Russia
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Anna Tvorogova
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| | - Vladimir Gontarenko
- Department of Vascular Surgery, National Medical Research Centre of Surgery Named after A.V. Vishnevsky under the RF Public Health Ministry, 117997 Moscow, Russia
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Cardiology Department, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127006 Moscow, Russia
- City Clinical Hospital Named after I.V. Davydovsky, Moscow Department of Healthcare, 109240 Moscow, Russia; (G.R.)
| |
Collapse
|
14
|
Waksman R, Merdler I, Case BC, Waksman O, Porto I. Targeting inflammation in atherosclerosis: overview, strategy and directions. EUROINTERVENTION 2024; 20:32-44. [PMID: 38165117 PMCID: PMC10756224 DOI: 10.4244/eij-d-23-00606] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a chronic condition characterised by the build-up of plaque in the inner lining of the blood vessels and it is the main underlying cause of cardiovascular disease. The development of atherosclerosis is associated with the accumulation of cholesterol and inflammation. Although effective therapies exist to lower low-density lipoprotein cholesterol (LDL-C) levels, some patients still experience cardiovascular events due to persistent inflammation, known as residual inflammatory risk (RIR). Researchers have conducted laboratory and animal studies to investigate the measurement and targeting of the inflammatory cascade associated with atherosclerosis, which have yielded promising results. In addition to guideline-directed lifestyle modifications and optimal medical therapy focusing on reducing LDL-C levels, pharmacological interventions targeting inflammation may provide further assistance in preventing future cardiac events. This review aims to explain the mechanisms of inflammation in atherosclerosis, identifies potential biomarkers, discusses available therapeutic options and their strengths and limitations, highlights future advancements, and summarises notable clinical studies. Finally, an evaluation and management algorithm for addressing RIR is presented.
Collapse
Affiliation(s)
- Ron Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Ilan Merdler
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Brian C Case
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Ori Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Italo Porto
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - Italian IRCCS Cardiology Network, Genoa, Italy
| |
Collapse
|
15
|
Burzynski LC, Morales-Maldonado A, Rodgers A, Kitt LA, Humphry M, Figg N, Bennett MR, Clarke MCH. Thrombin-activated interleukin-1α drives atherogenesis, but also promotes vascular smooth muscle cell proliferation and collagen production. Cardiovasc Res 2023; 119:2179-2189. [PMID: 37309666 PMCID: PMC10578913 DOI: 10.1093/cvr/cvad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 06/14/2023] Open
Abstract
AIMS Atherosclerosis is driven by multiple processes across multiple body systems. For example, the innate immune system drives both atherogenesis and plaque rupture via inflammation, while coronary artery-occluding thrombi formed by the coagulation system cause myocardial infarction and death. However, the interplay between these systems during atherogenesis is understudied. We recently showed that coagulation and immunity are fundamentally linked by the activation of interleukin-1α (IL-1α) by thrombin, and generated a novel knock-in mouse in which thrombin cannot activate endogenous IL-1α [IL-1α thrombin mutant (IL-1αTM)]. METHODS AND RESULTS Here, we show significantly reduced atherosclerotic plaque formation in IL-1αTM/Apoe-/- mice compared with Apoe-/- and reduced T-cell infiltration. However, IL-1αTM/Apoe-/- plaques have reduced vascular smooth muscle cells, collagen, and fibrous caps, indicative of a more unstable phenotype. Interestingly, the reduced atherogenesis seen with thrombin inhibition was absent in IL-1αTM/Apoe-/- mice, suggesting that thrombin inhibitors can affect atherosclerosis via reduced IL-1α activation. Finally, bone marrow chimeras show that thrombin-activated IL-1α is derived from both vessel wall and myeloid cells. CONCLUSIONS Together, we reveal that the atherogenic effect of ongoing coagulation is, in part, mediated via thrombin cleavage of IL-1α. This not only highlights the importance of interplay between systems during disease and the potential for therapeutically targeting IL-1α and/or thrombin, but also forewarns that IL-1 may have a role in plaque stabilization.
Collapse
Affiliation(s)
- Laura C Burzynski
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Alejandra Morales-Maldonado
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Amanda Rodgers
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Lauren A Kitt
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Melanie Humphry
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Nichola Figg
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Martin R Bennett
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Murray C H Clarke
- Section of CardioRespiratory Medicine, The Heart and Lung Research
Institute, The University of Cambridge, Papworth Road,
Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| |
Collapse
|
16
|
Jing J, Guo J, Dai R, Zhu C, Zhang Z. Targeting gut microbiota and immune crosstalk: potential mechanisms of natural products in the treatment of atherosclerosis. Front Pharmacol 2023; 14:1252907. [PMID: 37719851 PMCID: PMC10504665 DOI: 10.3389/fphar.2023.1252907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory reaction that primarily affects large and medium-sized arteries. It is a major cause of cardiovascular disease and peripheral arterial occlusive disease. The pathogenesis of AS involves specific structural and functional alterations in various populations of vascular cells at different stages of the disease. The immune response is involved throughout the entire developmental stage of AS, and targeting immune cells presents a promising avenue for its treatment. Over the past 2 decades, studies have shown that gut microbiota (GM) and its metabolites, such as trimethylamine-N-oxide, have a significant impact on the progression of AS. Interestingly, it has also been reported that there are complex mechanisms of action between GM and their metabolites, immune responses, and natural products that can have an impact on AS. GM and its metabolites regulate the functional expression of immune cells and have potential impacts on AS. Natural products have a wide range of health properties, and researchers are increasingly focusing on their role in AS. Now, there is compelling evidence that natural products provide an alternative approach to improving immune function in the AS microenvironment by modulating the GM. Natural product metabolites such as resveratrol, berberine, curcumin, and quercetin may improve the intestinal microenvironment by modulating the relative abundance of GM, which in turn influences the accumulation of GM metabolites. Natural products can delay the progression of AS by regulating the metabolism of GM, inhibiting the migration of monocytes and macrophages, promoting the polarization of the M2 phenotype of macrophages, down-regulating the level of inflammatory factors, regulating the balance of Treg/Th17, and inhibiting the formation of foam cells. Based on the above, we describe recent advances in the use of natural products that target GM and immune cells crosstalk to treat AS, which may bring some insights to guide the treatment of AS.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Dai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chaojun Zhu
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
17
|
Hinkley H, Counts DA, VonCanon E, Lacy M. T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease. Cells 2023; 12:2152. [PMID: 37681883 PMCID: PMC10486666 DOI: 10.3390/cells12172152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we discuss the recognition of atherosclerosis-related antigens that may trigger T cell activation together with additional signaling from co-stimulatory molecules and lesional cytokines. Although few studies have indicated candidates for the antigen specificity of T cells in atherosclerosis, further research is needed. Furthermore, we describe the pro-atherogenic and atheroprotective roles of diverse subsets of T cells such as CD4+ helper, CD8+ cytotoxic, invariant natural killer, and γδ T cells. To classify and quantify T cell subsets in atherosclerosis, we summarize current methods to analyze cellular heterogeneity including single cell RNA sequencing and T cell receptor (TCR) sequencing. Further insights into T cell biology will help shed light on the immunopathology of atherosclerosis, inform potential therapeutic interventions, and pave the way for precision medicine approaches in combating cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Michael Lacy
- Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
18
|
Głuchowska A, Cysewski D, Baj-Krzyworzeka M, Szatanek R, Węglarczyk K, Podszywałow-Bartnicka P, Sunderland P, Kozłowska E, Śliwińska MA, Dąbrowski M, Sikora E, Mosieniak G. Unbiased proteomic analysis of extracellular vesicles secreted by senescent human vascular smooth muscle cells reveals their ability to modulate immune cell functions. GeroScience 2022; 44:2863-2884. [PMID: 35900662 PMCID: PMC9768090 DOI: 10.1007/s11357-022-00625-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/12/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis, a common age-related disease, is characterized by intense immunological activity. Atherosclerotic plaque is composed of endothelial cells, vascular smooth muscle cells (VSMCs), lipids and immune cells infiltrating from the blood. During progression of the disease, VSMCs undergo senescence within the plaque and secrete SASP (senescence-associated secretory phenotype) factors that can actively modulate plaque microenvironment. We demonstrated that senescent VSMCs secrete increased number of extracellular vesicles (senEVs). Based on unbiased proteomic analysis of VMSC-derived EVs and of the soluble fraction of SASP (sSASP), more than 900 proteins were identified in each of SASP compartments. Comparison of the composition of VMSC-derived EVs with the SASP atlas revealed several proteins, including Serpin Family F Member 1 (SERPINF1) and Thrombospondin 1 (THBS1), as commonly upregulated components of EVs secreted by senescent VSMCs and fibroblasts. Among soluble SASP factors, only Growth Differentiation Factor 15 (GDF15) was universally increased in the secretome of senescent VSMCs, fibroblasts, and epithelial cells. Bioinformatics analysis of EV proteins distinguished functionally organized protein networks involved in immune cell function regulation. Accordingly, EVs released by senescent VSMCs induced secretion of IL-17, INFγ, and IL-10 by T cells and of TNFα produced by monocytes. Moreover senEVs influenced differentiation of monocytes favoring mix M1/M2 polarization with proinflammatory characteristics. Altogether, our studies provide a complex, unbiased analysis of VSMC SASP and prove that EVs derived from senescent VSMCs influence the cytokine milieu by modulating immune cell activity. Our results strengthen the role of senescent cells as an important inducer of inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Agata Głuchowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Clinical Research Centre, Medical University of Bialystok, Białystok, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Rafał Szatanek
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | | | - Piotr Sunderland
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Ewa Kozłowska
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology University of Warsaw, Warsaw, Poland
| | - Małgorzata A Śliwińska
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Grażyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland.
| |
Collapse
|
19
|
Toribio M, Wilks MQ, Hedgire S, Lu MT, Cetlin M, Wang M, Alhallak I, Durbin CG, White KS, Wallis Z, Schnittman SR, Stanley TL, El-Fakhri G, Lee H, Autissier P, Zanni MV, Williams KC, Grinspoon SK. Increased Macrophage-Specific Arterial Infiltration Relates to Noncalcified Plaque and Systemic Immune Activation in People With Human Immunodeficiency Virus. J Infect Dis 2022; 226:1823-1833. [PMID: 35856671 PMCID: PMC10205602 DOI: 10.1093/infdis/jiac301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Persistent immune activation is thought to contribute to heightened atherosclerotic cardiovascular disease (ASCVD) risk among people with human immunodeficiency virus (PWH). METHODS Participants (≥18 years) with or without human immunodeficiency virus (HIV) and without history of clinical ASCVD were enrolled. We hypothesized that increased macrophage-specific arterial infiltration would relate to plaque composition and systemic immune activation among PWH. We applied a novel targeted molecular imaging approach (technetium-99m [99mTc]-tilmanocept single photon emission computed tomography [SPECT]/CT) and comprehensive immune phenotyping. RESULTS Aortic 99mTc-tilmanocept uptake was significantly higher among PWH (n = 20) than participants without HIV (n = 10) with similar 10-year ASCVD risk (P = .02). Among PWH, but not among participants without HIV, noncalcified aortic plaque volume related directly to aortic 99mTc-tilmanocept uptake at different uptake thresholds. An interaction (P = .001) was seen between HIV status and noncalcified plaque volume, but not calcified plaque (P = .83). Systemic levels of caspase-1 (P = .004), CD14-CD16+ (nonclassical/patrolling/homing) monocytes (P = .0004) and CD8+ T cells (P = .005) related positively and CD4+/CD8+ T-cell ratio (P = .02) inversely to aortic 99mTc-tilmanocept uptake volume. CONCLUSIONS Macrophage-specific arterial infiltration was higher among PWH and related to noncalcified aortic plaque volume only among PWH. Key systemic markers of immune activation relating to macrophage-specific arterial infiltration may contribute to heightened ASCVD risk among PWH. CLINICAL TRIALS REGISTRATION NCT02542371.
Collapse
Affiliation(s)
- Mabel Toribio
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Moses Q Wilks
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sandeep Hedgire
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Madeline Cetlin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Melissa Wang
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Iad Alhallak
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Claudia G Durbin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin S White
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Zoey Wallis
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Samuel R Schnittman
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Georges El-Fakhri
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Autissier
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Potashnikova DM, Saidova AA, Tvorogova AV, Anisimova AS, Botsina AY, Vasilieva EY, Margolis LB. CTLs From Patients With Atherosclerosis Show Elevated Adhesiveness and Distinct Integrin Expression Patterns on 2D Substrates. Front Med (Lausanne) 2022; 9:891916. [PMID: 35911408 PMCID: PMC9328274 DOI: 10.3389/fmed.2022.891916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is the major cause of cardiovascular disease that is characterized by plaque formation in the blood vessel wall. Atherosclerotic plaques represent sites of chronic inflammation with diverse cell content that is shifted toward the prevalence of cytotoxic T-lymphocytes (CTLs) upon plaque progression. The studies of CTL recruitment to atherosclerotic plaques require adequate in vitro models accounting for CTL interactions with chemokine-ligands and extracellular matrix fibers via surface chemokine receptors and integrins. Here we applied such a model by investigating CTL adhesion and migration on six types of coated surfaces. We assessed adhesion and motility metrics, the expression of chemokine receptors, and integrins in CTLs of patients with atherosclerosis and healthy donors. Using fibronectin, platelet-poor plasma from patients with atherosclerosis, and conditioned medium from atherosclerotic plaques we revealed the role of substrate in CTL adhesiveness: fibronectin alone and fibronectin combined with platelet-poor plasma and conditioned medium elevated the CTL adhesiveness - in patients the elevation was significantly higher than in healthy donors (p = 0.02, mixed 2-way ANOVA model). This was in line with our finding that the expression levels of integrin-coding mRNAs were elevated in the presence of fibronectin (p < 0.05) and ITGB1, ITGA1, and ITGA4 were specifically upregulated in patients compared to healthy donors (p < 0.01). Our experimental model did not affect the expression levels of mRNAs CCR4, CCR5, and CX3CR1 coding the chemokine receptors that drive T-lymphocyte migration to plaques. Thus, we demonstrated the substrate-dependence of integrin expression and discriminated CTLs from patients and healthy donors by adhesion parameters and integrin expression levels.
Collapse
Affiliation(s)
- Daria M. Potashnikova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Aleena A. Saidova
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Anna V. Tvorogova
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Alexandra S. Anisimova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Alexandra Yu Botsina
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Elena Yu Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Moscow Department of Healthcare, City Clinical Hospital Named After I.V. Davydovsky, Moscow, Russia
| | - Leonid B. Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Khare HA, Døssing KBV, Ringgaard L, Christensen E, Urbak L, Sillesen H, Ripa RS, Binderup T, Pedersen SF, Kjaer A. In vivo detection of urokinase-type plasminogen activator receptor (uPAR) expression in arterial atherogenesis using [64Cu]Cu-DOTA-AE105 positron emission tomography (PET). Atherosclerosis 2022; 352:103-111. [DOI: 10.1016/j.atherosclerosis.2022.03.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
|
22
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
23
|
Oh KK, Adnan M, Cho DH. A network pharmacology analysis on drug-like compounds from Ganoderma lucidum for alleviation of atherosclerosis. J Food Biochem 2021; 45:e13906. [PMID: 34409623 DOI: 10.1111/jfbc.13906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/26/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Ganoderma lucidum (GL) is known as a potent alleviator against chronic inflammatory disease like atherosclerosis (AS), but its mechanisms against AS have not been unveiled. This research aimed to identify the key compounds(s) and mechanism(s) of GL against AS through network pharmacology. The compounds from GL were identified by gas chromatography-mass spectrum (GC-MS), and SwissADME screened their physicochemical properties. Then, the target(s) associated with the screened compound(s) or AS related targets were identified by public databases, and we selected the overlapping targets using a Venn diagram. The networks between overlapping targets and compounds were visualized, constructed, and analyzed by RStudio. Finally, we performed a molecular docking test (MDT) to explore key target(s), compound(s), on AutoDockVina. A total of 35 compounds in GL were detected via GC-MS, and 34 compounds (accepted by Lipinski's rule) were selected as drug-like compounds (DLCs). A total of 34 compounds were connected to the number of 785 targets, and DisGeNET and Online Mendelian Inheritance in Man (OMIM) identified 2,606 AS-related targets. The final 98 overlapping targets were extracted between the compounds-targets and AS-related targets. On Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, the number of 27 signaling pathways were sorted out, and a hub signaling pathway (MAPK signaling pathway), a core gene (PRKCA), and a key compound (Benzamide, 4-acetyl-N-[2,6-dimethylphenyl]) were selected among the 27 signaling pathways via MDT. Overall, we found that the identified 3 DLCs from GL have potent anti-inflammatory efficacy, improving AS by inactivating the MAPK signaling pathway. PRACTICAL APPLICATIONS: Ganoderma lucidum (GL) has been used as a medicinal or edible mushroom for chronic inflammatory patients: diabetes mellitus and dyslipidemia, especially atherosclerosis (AS). Until now, the majority of mushroom research has been implemented regarding β-glucan derivatives with very hydrophilic physicochemical properties. It implies that β-glucan or its derivatives have poor bioavailability. Hence, we have involved GC-MS in identifying lipophilic compounds from GL, which filtered them in silico to sort drug-like compounds (DLCs). Then, we retrieved targets associated with the DLCs, and identified a key signaling pathway, key targets, and key compounds against AS. In this paper, we utilized bioinformatics and network pharmacology theory to understand the uncovered pharmacological mechanism of GL on AS. To sum things up, our analysis elucidates the relationships between signaling pathways, targets, and compounds in GL. Ultimately, this work provides biochemical evidence to identify the therapeutic effect of GL on AS, and a scientific basis for deciphering the key mechanism on DLCs of GL against AS.
Collapse
Affiliation(s)
- Ki Kwang Oh
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Korea
| | - Md Adnan
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Korea
| | - Dong Ha Cho
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
24
|
Hill CA, Fernandez DM, Giannarelli C. Single cell analyses to understand the immune continuum in atherosclerosis. Atherosclerosis 2021; 330:85-94. [PMID: 33934886 PMCID: PMC8855459 DOI: 10.1016/j.atherosclerosis.2021.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is initiated by the accumulation of lipids in the arterial wall that trigger a complex and poorly understood network of inflammatory processes. At the same time, recent clinical findings reveal that targeting specific immune alterations in patients with cardiovascular disease (CVD) represents a promising approach to preventing recurrent cardiovascular events. In order to achieve these tailored therapies, it is critical to resolve the heterogenous environment of the atherosclerotic lesion and decipher the complex structural and functional changes which immune cells undergo throughout disease progression. Recently, single-cell approaches including single cell mass cytometry by time of flight (CyTOF), single cell RNA sequencing (scRNA-seq) and Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) have emerged as valuable tools in resolving cellular plasticity within atherosclerotic lesions. In this review, we will discuss the most important insights that have been gleaned from the application of these single-cell approaches to validated experimental models of atherosclerosis. Additionally, as clinical progress in treatment of the disease depends on the translation of discoveries to human tissues, we will also examine the challenges associated with the application of single-cell approaches to human vascular tissue and the discoveries made by the initial efforts in this direction. Finally, we will analyze the advantages and limitations of dissociative single-cell approaches and how novel in-situ technologies could advance the field by allowing for the investigation of individual cells while preserving the heterogenous architecture of the atherosclerotic lesion.
Collapse
Affiliation(s)
| | | | - Chiara Giannarelli
- Department of Medicine, Cardiovascular Research Center, USA; Department of Genetics and Genomic Sciences, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Cardiology and Cardiovascular Research Center at NYU Langone NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Pakzad B, Rajae E, Shahrabi S, Mansournezhad S, Davari N, Azizidoost S, Saki N. T-Cell Molecular Modulation Responses in Atherosclerosis Anergy. Lab Med 2021; 51:557-565. [PMID: 32106301 DOI: 10.1093/labmed/lmaa003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis continues to be a major cause of death in patients with cardiovascular diseases. The cooperative role of immunity has been recently considered in atherosclerotic plaque inflammation, especially adaptive immune response by T cells. In this review, we examine the possible role of T cells in atherosclerosis-mediated inflammation and conceivable therapeutic strategies that can ameliorate complications of atherosclerosis. The cytokines secreted by T-lymphocyte subsets, different pathophysiological profiles of microRNAs (miRs), and the growth factor/receptor axis have diverse effects on the inflammatory cycle of atherosclerosis. Manipulation of miRNA expression and prominent growth factor receptors involved in inflammatory cytokine secretion in atherosclerosis can be considered diagnostic biomarkers in the induction of anergy and blockade of atherosclerotic development. This manuscript reviews immunomodulation of T cells responses in atherosclerosis anergy.
Collapse
Affiliation(s)
- Bahram Pakzad
- Internal Medicine Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Rajae
- Department of Rheumatology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- -Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Somayeh Mansournezhad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
26
|
Das D, Podder S. Unraveling the molecular crosstalk between Atherosclerosis and COVID-19 comorbidity. Comput Biol Med 2021; 134:104459. [PMID: 34020127 PMCID: PMC8088080 DOI: 10.1016/j.compbiomed.2021.104459] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Corona virus disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus -2 (SARS-CoV-2) has created ruckus throughout the world. Growing epidemiological studies have depicted atherosclerosis as a comorbid factor of COVID-19. Though both these diseases are triggered via inflammatory rage that leads to injury of healthy tissues, the molecular linkage between them and their co-influence in causing fatality is not yet understood. METHODS We have retrieved the data of differentially expressed genes (DEGs) for both atherosclerosis and COVID-19 from publicly available microarray and RNA-Seq datasets. We then reconstructed the protein-protein interaction networks (PPIN) for these diseases from protein-protein interaction data of corresponding DEGs. Using RegNetwork and TRRUST, we mapped the transcription factors (TFs) in atherosclerosis and their targets (TGs) in COVID-19 PPIN. RESULTS From the atherosclerotic PPIN, we have identified 6 hubs (TLR2, TLR4, EGFR, SPI1, MYD88 and IRF8) as differentially expressed TFs that might control the expression of their 17 targets in COVID-19 PPIN. The important target proteins include IL1B, CCL5, ITGAM, IFIT3, CXCL1, CXCL2, CXCL3 and CXCL8. Consequent functional enrichment analysis of these TGs have depicted inflammatory responses to be overrepresented among the gene sets. CONCLUSION Finally, analyzing the DEGs in cardiomyocytes infected with SARS-CoV-2, we have concluded that MYD88 is a crucial linker of atherosclerosis and COVID-19, the co-existence of which lead to fatal outcomes. Anti-inflammatory therapy targeting MYD88 could be a potent strategy for combating this comorbidity.
Collapse
Affiliation(s)
- Deepyaman Das
- Department of Microbiology, Raiganj University, Raiganj, Uttar Dinajpur, 733134, West Bengal, India
| | - Soumita Podder
- Department of Microbiology, Raiganj University, Raiganj, Uttar Dinajpur, 733134, West Bengal, India.
| |
Collapse
|
27
|
Infante T, Franzese M, Ruocco A, Schiano C, Affinito O, Pane K, Memoli D, Rizzo F, Weisz A, Bontempo P, Grimaldi V, Berrino L, Soricelli A, Mauro C, Napoli C. ABCA1, TCF7, NFATC1, PRKCZ, and PDGFA DNA methylation as potential epigenetic-sensitive targets in acute coronary syndrome via network analysis. Epigenetics 2021; 17:547-563. [PMID: 34151742 DOI: 10.1080/15592294.2021.1939481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Acute coronary syndrome (ACS) is the most severe clinical manifestation of coronary heart disease.We performed an epigenome-wide analysis of circulating CD4+ and CD8+ T cells isolated from ACS patients and healthy subjects (HS), enrolled in the DIANA clinical trial, by reduced-representation bisulphite sequencing (RRBS). In CD4+ T cells, we identified 61 differentially methylated regions (DMRs) associated with 57 annotated genes (53% hyper- and 47% hypo-methylated) by comparing ACS patients vs HS. In CD8+ T cells, we identified 613 DMRs associated with 569 annotated genes (28% hyper- and 72% hypo-methylated) in ACS patients as compared to HS. In CD4+ vs CD8+ T cells of ACS patients we identified 175 statistically significant DMRs associated with 157 annotated genes (41% hyper- and 59% hypo-methylated). From pathway analyses, we selected six differentially methylated hub genes (NFATC1, TCF7, PDGFA, PRKCB, PRKCZ, ABCA1) and assessed their expression levels by q-RT-PCR. We found an up-regulation of selected genes in ACS patients vs HS (P < 0.001). ABCA1, TCF7, PDGFA, and PRKCZ gene expression was positively associated with CK-MB serum concentrations (r = 0.75, P = 0.03; r = 0.760, P = 0.029; r = 0.72, P = 0.044; r = 0.74, P = 0.035, respectively).This pilot study is the first single-base resolution map of DNA methylome by RRBS in CD4+ and CD8+ T cells and provides specific methylation signatures to clarify the role of aberrant methylation in ACS pathogenesis, thus supporting future research for novel epigenetic-sensitive biomarkers in the prevention and early diagnosis of this pathology.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Antonio Ruocco
- Unit of Cardiovascular Diseases and Arrhythmias, "Antonio Cardarelli" Hospital, Naples, Italy
| | - Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | - Domenico Memoli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Francesca Rizzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Alessandro Weisz
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Paola Bontempo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Grimaldi
- IRCCS SDN, Naples, Italy.,U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Soricelli
- IRCCS SDN, Naples, Italy.,Department of Exercise and Wellness Sciences, University of Naples Parthenope, Naples, Italy
| | - Ciro Mauro
- Unit of Cardiovascular Diseases and Arrhythmias, "Antonio Cardarelli" Hospital, Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
28
|
Kuan R, Agrawal DK, Thankam FG. Treg cells in atherosclerosis. Mol Biol Rep 2021; 48:4897-4910. [PMID: 34117978 DOI: 10.1007/s11033-021-06483-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022]
Abstract
Atherosclerosis involves both innate and adaptive immunity. Here, we provide an overview of the role of regulatory T (Treg) cells in atherosclerotic diseases. Treg cells and their inhibitory cytokines, IL-10 and TGF-β, have been identified in atherosclerotic lesions and to inhibit progression through lipoprotein metabolism modulation. Treg cells have also been found to convert to T follicular helper (Tfh) cells and promote atherosclerosis progression. Treg cell involvement in different stages of atherosclerotic progression and Treg cell-mediated modulation of plaque development occurs via inflammation suppression and atheroma formation has been focused. Moreover, existing knowledge suggests that Treg cells are likely involved in the pathology of other specific circumstances including in-stent restenosis, neointimal hyperplasia, vessel graft failure, and ischemic arterial injury; however, there remain gaps regarding their specific contribution. Hence, advancements in the knowledge regarding Treg cells in diverse aspects of atherosclerosis offer translational significance for the management of atherosclerosis and associated diseases.
Collapse
Affiliation(s)
- Rebecca Kuan
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
29
|
Leistner DM, Kränkel N, Meteva D, Abdelwahed YS, Seppelt C, Stähli BE, Rai H, Skurk C, Lauten A, Mochmann HC, Fröhlich G, Rauch-Kröhnert U, Flores E, Riedel M, Sieronski L, Kia S, Strässler E, Haghikia A, Dirks F, Steiner JK, Mueller DN, Volk HD, Klotsche J, Joner M, Libby P, Landmesser U. Differential immunological signature at the culprit site distinguishes acute coronary syndrome with intact from acute coronary syndrome with ruptured fibrous cap: results from the prospective translational OPTICO-ACS study. Eur Heart J 2021; 41:3549-3560. [PMID: 33080003 DOI: 10.1093/eurheartj/ehaa703] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/15/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
AIMS Acute coronary syndromes with intact fibrous cap (IFC-ACS), i.e. caused by coronary plaque erosion, account for approximately one-third of ACS. However, the underlying pathophysiological mechanisms as compared with ACS caused by plaque rupture (RFC-ACS) remain largely undefined. The prospective translational OPTICO-ACS study programme investigates for the first time the microenvironment of ACS-causing culprit lesions (CL) with intact fibrous cap by molecular high-resolution intracoronary imaging and simultaneous local immunological phenotyping. METHODS AND RESULTS The CL of 170 consecutive ACS patients were investigated by optical coherence tomography (OCT) and simultaneous immunophenotyping by flow cytometric analysis as well as by effector molecule concentration measurements across the culprit lesion gradient (ratio local/systemic levels). Within the study cohort, IFC caused 24.6% of ACS while RFC-ACS caused 75.4% as determined and validated by two independent OCT core laboratories. The IFC-CL were characterized by lower lipid content, less calcification, a thicker overlying fibrous cap, and largely localized near a coronary bifurcation as compared with RFC-CL. The microenvironment of IFC-ACS lesions demonstrated selective enrichment in both CD4+ and CD8+ T-lymphocytes (+8.1% and +11.2%, respectively, both P < 0.05) as compared with RFC-ACS lesions. T-cell-associated extracellular circulating microvesicles (MV) were more pronounced in IFC-ACS lesions and a significantly higher amount of CD8+ T-lymphocytes was detectable in thrombi aspirated from IFC-culprit sites. Furthermore, IFC-ACS lesions showed increased levels of the T-cell effector molecules granzyme A (+22.4%), perforin (+58.8%), and granulysin (+75.4%) as compared with RFC plaques (P < 0.005). Endothelial cells subjected to culture in disturbed laminar flow conditions, i.e. to simulate coronary flow near a bifurcation, demonstrated an enhanced adhesion of CD8+T cells. Finally, both CD8+T cells and their cytotoxic effector molecules caused endothelial cell death, a key potential pathophysiological mechanism in IFC-ACS. CONCLUSIONS The OPTICO-ACS study emphasizes a novel mechanism in the pathogenesis of IFC-ACS, favouring participation of the adaptive immune system, particularly CD4+ and CD8+ T-cells and their effector molecules. The different immune signatures identified in this study advance the understanding of coronary plaque progression and may provide a basis for future development of personalized therapeutic approaches to ACS with IFC. TRIAL REGISTRATION The study was registered at clinicalTrials.gov (NCT03129503).
Collapse
Affiliation(s)
- David M Leistner
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Nicolle Kränkel
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Denitsa Meteva
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Youssef S Abdelwahed
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Claudio Seppelt
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Barbara E Stähli
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Himanshu Rai
- DZHK (German Centre for Cardiovascular Research) Partner Site Munch, Munich, 80636, Germany
| | - Carsten Skurk
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Alexander Lauten
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Hans-Christian Mochmann
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany
| | - Georg Fröhlich
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Ursula Rauch-Kröhnert
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Eduardo Flores
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany
| | - Matthias Riedel
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Lara Sieronski
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Sylvia Kia
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Elisabeth Strässler
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Arash Haghikia
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Fabian Dirks
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| | - Julia K Steiner
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany
| | - Dominik N Mueller
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany.,Experimental and Clinical Research Centre (ECRC), a cooperation of Charité University Medicine Berlin and Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health (BIH), Berlin 10117, Germany.,Institute for Medical Immunology and BIH Centre for Regenerative Therapies (BCRT), and Berlin Centre for Advanced Therapies (BeCAT), Charité University Medicine Berlin, Berlin 13353, Germany
| | - Jens Klotsche
- German Rheumatism Research Centre Berlin, and Institute for Social Medicine, Epidemiology und Heath Economy, Charité University Medicine Berlin, Campus Charité Mitte, Berlin 10117, Germany
| | - Michael Joner
- Department of Cardiology and ISAR Research Centre, German Heart Centre, Munich, 80636, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Munch, Munich, 80636, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Ave Louis Pasteur, Boston, MA 02115, USA
| | - Ulf Landmesser
- Department of Cardiology, University Heart Centre Berlin and Charité University Medicine Berlin, Campus Benjamin-Franklin (CBF), Hindenburgdamm 30, Berlin D-12203, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin 12203, Germany.,Berlin Institute of Health (BIH), Berlin 10117, Germany
| |
Collapse
|
30
|
Abstract
Atherosclerotic lesions are populated by cells of the innate and adaptive immune system, including CD8+ T cells. The CD8+ T cell infiltrate has recently been characterized in mouse and human atherosclerosis and revealed activated, cytotoxic, and possibly dysfunctional and exhausted cell phenotypes. In mouse models of atherosclerosis, antibody-mediated depletion of CD8+ T cells ameliorates atherosclerosis. CD8+ T cells control monopoiesis and macrophage accumulation in early atherosclerosis. In addition, CD8+ T cells exert cytotoxic functions in atherosclerotic plaques and contribute to macrophage cell death and necrotic core formation. CD8+ T cell activation may be antigen-specific, and epitopes of atherosclerosis-relevant antigens may be targets of CD8+ T cells and their cytotoxic activity. CD8+ T cell functions are tightly controlled by costimulatory and coinhibitory immune checkpoints. Subsets of regulatory CD25+CD8+ T cells with immunosuppressive functions can inhibit atherosclerosis. Importantly, local cytotoxic CD8+ T cell responses may trigger endothelial damage and plaque erosion in acute coronary syndromes. Understanding the complex role of CD8+ T cells in atherosclerosis may pave the way for defining novel treatment approaches in atherosclerosis. In this review article, we discuss these aspects, highlighting the emerging and critical role of CD8+ T cells in atherosclerosis.
Collapse
|
31
|
Adaptive Immune Responses in Human Atherosclerosis. Int J Mol Sci 2020; 21:ijms21239322. [PMID: 33297441 PMCID: PMC7731312 DOI: 10.3390/ijms21239322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that is initiated by the deposition and accumulation of low-density lipoproteins in the artery wall. In this review, we will discuss the role of T- and B-cells in human plaques at different stages of atherosclerosis and the utility of profiling circulating immune cells to monitor atherosclerosis progression. Evidence supports a proatherogenic role for intraplaque T helper type 1 (Th1) cells, CD4+CD28null T-cells, and natural killer T-cells, whereas Th2 cells and regulatory T-cells (Treg) have an atheroprotective role. Several studies indicate that intraplaque T-cells are activated upon recognition of endogenous antigens including heat shock protein 60 and oxidized low-density lipoprotein, but antigens derived from pathogens can also trigger T-cell proliferation and cytokine production. Future studies are needed to assess whether circulating cellular biomarkers can improve identification of vulnerable lesions so that effective intervention can be implemented before clinical manifestations are apparent.
Collapse
|
32
|
Padgett LE, Dinh HQ, Wu R, Gaddis DE, Araujo DJ, Winkels H, Nguyen A, McNamara CA, Hedrick CC. Naive CD8 + T Cells Expressing CD95 Increase Human Cardiovascular Disease Severity. Arterioscler Thromb Vasc Biol 2020; 40:2845-2859. [PMID: 33054398 DOI: 10.1161/atvbaha.120.315106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) remains a significant global health concern with a high degree of mortality. While CD4+ T cells have been extensively studied in CVD, the importance of CD8+ T cells in this disease, despite their abundance and increased activation in human atherosclerotic plaques, remains largely unknown. Thus, the objective of this study was to compare peripheral T-cell signatures between humans with a high (severe) risk of CVD (including myocardial infarction or stroke) and those with a low risk of CVD. Approach and Results: Using mass cytometry, we uncovered a naive CD8+ T (TN) cell population expressing CD95 (termed CD95+CD8+ stem cell memory T [CD8 TSCM] cells) that was enriched in patients with high compared with low CVD. This T-cell subset enrichment within individuals with high CVD was a relative increase and resulted from the loss of CD95lo cells within the TN compartment. We found that CD8 TSCM cells positively correlated with CVD risk in humans, while CD8+ TN cells were inversely correlated. Atherosclerotic apolipoprotein E-deficient (ApoE-/-) mice also displayed respective 7- and 2-fold increases in CD8+ TSCM frequencies within the peripheral blood and aorta-draining paraaortic lymph nodes compared with C57BL/6J mice. CD8+ TSCM cells were 1.7-fold increased in aortas from western diet fed ApoE-/- mice compared with normal laboratory diet-fed ApoE-/- mice. Importantly, transfer of TSCM cells into immune-deficient Rag.Ldlr recipient mice that lacked T cells increased atherosclerosis, illustrating the importance of these cells in atherogenesis. CONCLUSIONS CD8+ TSCM cells are increased in humans with high CVD. As these TSCM cells promote atherosclerosis, targeting them may attenuate atherosclerotic plaque progression.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Huy Q Dinh
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Runpei Wu
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Dalia E Gaddis
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Daniel J Araujo
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Holger Winkels
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Anh Nguyen
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Coleen A McNamara
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| |
Collapse
|
33
|
Rai A, Narisawa M, Li P, Piao L, Li Y, Yang G, Cheng XW. Adaptive immune disorders in hypertension and heart failure: focusing on T-cell subset activation and clinical implications. J Hypertens 2020; 38:1878-1889. [PMID: 32890260 DOI: 10.1097/hjh.0000000000002456] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
: Hypertension is a growing health concern worldwide. Established hypertension is a causative factor of heart failure, which is characterized by increased vascular resistance and intractable uncontrolled blood pressure. Hypertension and heart failure have multiple causes and complex pathophysiology but cellular immunity is thought to contribute to the development of both. Recent studies showed that T cells play critical roles in hypertension and heart failure in humans and animals, with various stimuli leading to the formation of effector T cells that infiltrate the cardiovascular wall. Monocytes/macrophages also accumulate in the cardiovascular wall. Various cytokines (e.g. interleukin-6, interleukin-17, interleukin-10, tumor necrosis factor-α, and interferon-γ) released from immune cells of various subtypes promote vascular senescence and elastic laminal degradation as well as cardiac fibrosis and/or hypertrophy, leading to cardiovascular structural alterations and dysfunction. Recent laboratory evidence has defined a link between inflammation and the immune system in initiation and progression of hypertension and heart failure. Moreover, cross-talk among natural killer cells, adaptive immune cells (T cells and B cells), and innate immune cells (i.e. monocytes, macrophages, neutrophils, and dendritic cells) contributes to end-cardiovasculature damage and dysfunction in hypertension and heart failure. Clinical and experimental studies on the diagnostic potential of T-cell subsets revealed that blood regulatory T cells, CD4 cells, CD8 T cells, and the ratio of CD4 to CD8 T cells show promise as biomarkers of hypertension and heart failure. Therapeutic interventions to suppress activation of these cells may prove beneficial in reducing end-organ damage and preventing consequences of cardiovascular failure, including hypertension of heart failure.
Collapse
Affiliation(s)
- Avinas Rai
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Limei Piao
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Yanglong Li
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Guang Yang
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital, Juzijie, Yanji, Jilin Province, China
| |
Collapse
|
34
|
Association of Major Histocompatibility Complex Class I Related Chain A/B Positive Microparticles with Acute Myocardial Infarction and Disease Severity. Diagnostics (Basel) 2020; 10:diagnostics10100766. [PMID: 33003303 PMCID: PMC7656305 DOI: 10.3390/diagnostics10100766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/23/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Various cell types undergo activation and stress during atherosclerosis resulting in the development of acute myocardial infarction (AMI) in coronary artery disease (CAD). Major histocompatibility complex class I related chain A and B (MICA/B) can be expressed on the surface of activated and stressed cells and released into blood circulation in several forms including microparticles (MICA/B+ MPs) from various cell types. We aimed to investigate the association of these MICA/B+ MPs with the presence of AMI. Fifty-one AMI and 46 age-matched control subjects were recruited. Methods: Levels of MICA/B+ MPs derived from various parent cells including endothelial cells, platelets, monocytes, neutrophils, and T lymphocytes were determined by flow cytometry. Results: The levels and proportion of MICA/B+ MPs from all types of cell origin were significantly increased in AMI patients compared to those of the controls. A multivariate regression model showed an independent association between MICA/B+ MPs and AMI (OR = 11.6; 95% CI = 2.8, 47.3). Interestingly, based on the disease severity, we found that the levels of MICA/B+ MPs were significantly elevated in the ST-segment elevation myocardial infarction (STEMI) compared to the non-STEMI (NSTEMI) patients. Moreover, an independent association of MICA/B+ MPs with the occurrence of STEMI was also demonstrated (OR = 4.1; 95% CI = 1.5, 16.7). Conclusions: These results suggest that MICA/B+ MPs are associated with AMI and disease severity. They may act as mediators contributing to the pathological process of AMI. Alternatively, they are the results of various cell activations contributing to AMI.
Collapse
|
35
|
Panigrahi S, Chen B, Fang M, Potashnikova D, Komissarov AA, Lebedeva A, Michaelson GM, Wyrick JM, Morris SR, Sieg SF, Paiardini M, Villinger FJ, Harth K, Kashyap VS, Cameron MJ, Cameron CM, Vasilieva E, Margolis L, Younes SA, Funderburg NT, Zidar DA, Lederman MM, Freeman ML. CX3CL1 and IL-15 Promote CD8 T cell chemoattraction in HIV and in atherosclerosis. PLoS Pathog 2020; 16:e1008885. [PMID: 32976527 PMCID: PMC7540902 DOI: 10.1371/journal.ppat.1008885] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/07/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains an important cause of morbidity in the general population and risk for ASCVD is increased approximately 2-fold in persons living with HIV infection (PLWH). This risk is linked to elevated CD8 T cell counts that are abundant in atherosclerotic plaques and have been implicated in disease pathogenesis yet the mechanisms driving T cell recruitment to and activation within plaques are poorly defined. Here we investigated the role of CD8 T cells in atherosclerosis in a non-human primate model of HIV infection and in the HIV-uninfected elderly; we sought to identify factors that promote the activation, function, and recruitment to endothelium of CX3CR1+ CD8 T cells. We measured elevated expression of CX3CL1 and IL-15, and increased CD8 T cell numbers in the aortas of rhesus macaques infected with SIV or SHIV, and demonstrated similar findings in atherosclerotic vessels of HIV-uninfected humans. We found that recombinant TNF enhanced the production and release of CX3CL1 and bioactive IL-15 from aortic endothelial cells, but not from aortic smooth muscle cells. IL-15 in turn promoted CX3CR1 surface expression on and TNF synthesis by CD8 T cells, and IL-15-treated CD8 T cells exhibited enhanced CX3CL1-dependent chemoattraction toward endothelial cells in vitro. Finally, we show that CD8 T cells in human atherosclerotic plaques have an activated, resident phenotype consistent with in vivo IL-15 and CX3CL1 exposure. In this report, we define a novel model of CD8 T cell involvement in atherosclerosis whereby CX3CL1 and IL-15 operate in tandem within the vascular endothelium to promote infiltration by activated CX3CR1+ memory CD8 T cells that drive further endothelial activation via TNF. We propose that these interactions are prevalent in aging and in PLWH, populations where circulating activated CX3CR1+ CD8 T cell numbers are often expanded.
Collapse
Affiliation(s)
- Soumya Panigrahi
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Bonnie Chen
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Mike Fang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Daria Potashnikova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Moscow State University, Moscow, Russia
| | - Alexey A. Komissarov
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Anna Lebedeva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Gillian M. Michaelson
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Jonathan M. Wyrick
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Stephen R. Morris
- Cleveland Louis Stokes Veterans Affairs Medical Center, Cleveland, OH, United States of America
| | - Scott F. Sieg
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, United States of America
| | - Karem Harth
- Harrington Heart & Vascular Institute, University Hospitals, Cleveland Medical Center/Case Western Reserve University, School of Medicine, Cleveland, OH, United States of America
| | - Vikram S. Kashyap
- Harrington Heart & Vascular Institute, University Hospitals, Cleveland Medical Center/Case Western Reserve University, School of Medicine, Cleveland, OH, United States of America
| | - Mark J. Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Cheryl M. Cameron
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States of America
| | - Souheil-Antoine Younes
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Nicholas T. Funderburg
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States of America
| | - David A. Zidar
- Cleveland Louis Stokes Veterans Affairs Medical Center, Cleveland, OH, United States of America
- Harrington Heart & Vascular Institute, University Hospitals, Cleveland Medical Center/Case Western Reserve University, School of Medicine, Cleveland, OH, United States of America
| | - Michael M. Lederman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| | - Michael L. Freeman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States of America
| |
Collapse
|
36
|
Poels K, Vos WG, Lutgens E, Seijkens TTP. E3 Ubiquitin Ligases as Immunotherapeutic Target in Atherosclerotic Cardiovascular Disease. Front Cardiovasc Med 2020; 7:106. [PMID: 32582770 PMCID: PMC7292335 DOI: 10.3389/fcvm.2020.00106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic low-grade inflammation drives atherosclerosis and despite optimal pharmacological treatment of classical cardiovascular risk factors, one third of the patients with atherosclerotic cardiovascular disease has elevated inflammatory biomarkers. Additional anti-inflammatory strategies to target this residual inflammatory cardiovascular risk are therefore required. T-cells are a dominant cell type in human atherosclerotic lesions. Modulation of T-cell activation is therefore a potential strategy to target inflammation in atherosclerosis. Ubiquitination is an important regulatory mechanism of T-cell activation and several E3 ubiquitin ligases, including casitas B-lineage lymphoma proto-oncogene B (Cbl-B), itchy homolog (Itch), and gene related to anergy in lymphocytes (GRAIL), function as a natural brake on T-cell activation. In this review we discuss recent insights on the role of Cbl-B, Itch, and GRAIL in atherosclerosis and explore the therapeutic potential of these E3 ubiquitin ligases in cardiovascular medicine.
Collapse
Affiliation(s)
- Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Winnie G Vos
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Internal Medicine, Amsterdam UMC, Location VUmc, VU University, Amsterdam, Netherlands.,Department of Hematology, Amsterdam UMC, Location VUmc, VU University, Amsterdam, Netherlands
| |
Collapse
|
37
|
Talepoor AG, Fouladseresht H, Khosropanah S, Doroudchi M. Immune-Inflammation in Atherosclerosis: A New Twist in an Old Tale. Endocr Metab Immune Disord Drug Targets 2020; 20:525-545. [DOI: 10.2174/1871530319666191016095725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022]
Abstract
Background and Objective:Atherosclerosis, a chronic and progressive inflammatory disease, is triggered by the activation of endothelial cells followed by infiltration of innate and adaptive immune cells including monocytes and T cells in arterial walls. Major populations of T cells found in human atherosclerotic lesions are antigen-specific activated CD4+ effectors and/or memory T cells from Th1, Th17, Th2 and Treg subsets. In this review, we will discuss the significance of T cell orchestrated immune inflammation in the development and progression of atherosclerosis.Discussion:Pathogen/oxidative stress/lipid induced primary endothelial wound cannot develop to a full-blown atherosclerotic lesion in the absence of chronically induced inflammation. While the primary inflammatory response might be viewed as a lone innate response, the persistence of such a profound response over time must be (and is) associated with diverse local and systemic T cell responses. The interplay between T cells and innate cells contributes to a phenomenon called immuneinflammation and has an impact on the progression and outcome of the lesion. In recent years immuneinflammation, an old term, has had a comeback in connecting the puzzle pieces of chronic inflammatory diseases.Conclusion:Taking one-step back and looking from afar at the players of immune-inflammation may help us provide a broader perspective of these complicated interactions. This may lead to the identification of new drug targets and the development of new therapies as well as preventative measures.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Chen B, Morris SR, Panigrahi S, Michaelson GM, Wyrick JM, Komissarov AA, Potashnikova D, Lebedeva A, Younes SA, Harth K, Kashyap VS, Vasilieva E, Margolis L, Zidar DA, Sieg SF, Shive CL, Funderburg NT, Gianella S, Lederman MM, Freeman ML. Cytomegalovirus Coinfection Is Associated with Increased Vascular-Homing CD57 + CD4 T Cells in HIV Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2722-2733. [PMID: 32229536 PMCID: PMC7315224 DOI: 10.4049/jimmunol.1900734] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.
Collapse
Affiliation(s)
- Bonnie Chen
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Stephen R Morris
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Soumya Panigrahi
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Gillian M Michaelson
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Jonathan M Wyrick
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Alexey A Komissarov
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Daria Potashnikova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
- Department of Cell Biology and Histology, School of Biology, Moscow State University, Moscow 119234, Russia
| | - Anna Lebedeva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Souheil-Antoine Younes
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Karem Harth
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Vikram S Kashyap
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Leonid Margolis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - David A Zidar
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Scott F Sieg
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Carey L Shive
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH 43210; and
| | - Sara Gianella
- Center for AIDS Research, Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Michael M Lederman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Michael L Freeman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106;
| |
Collapse
|
39
|
Albiero R, Seresini G. Atherosclerotic spontaneous coronary artery dissection (A-SCAD) in a patient with COVID-19: case report and possible mechanisms. EUROPEAN HEART JOURNAL-CASE REPORTS 2020; 4:1-6. [PMID: 33089040 PMCID: PMC7239234 DOI: 10.1093/ehjcr/ytaa133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022]
Abstract
Background Spontaneous coronary artery dissection (SCAD) may be atherosclerotic (A-SCAD) or non-atherosclerotic (NA-SCAD) in origin. Contemporary usage of the term ‘SCAD’ is typically synonymous with NA-SCAD. COVID-19 could induce a vascular inflammation localized in the coronary adventitia and periadventitial fat and contribute to the development of an A-SCAD of a vulnerable plaque in a susceptible patient. Case summary In this report we describe a case of a COVID-19 patient with past cardiac history of CAD who was admitted for acute coronary syndrome (ACS). Coronary angiography demonstrated the culprit lesion in the proximal LAD that presented with a very complex and unusual morphology, indicative of an A-SCAD. The diagnosis of A-SCAD was supported by the presence of a mild stenosis in the same coronary segment in the last angiogram performed 3 years previously. He was successfully treated by PCI, had a favourable course of the COVID-19 with no symptoms of pneumonia, and was discharged from the hospital after two negative tests for SARS-CoV-2. Discussion A higher index of suspicion of A-SCAD is needed in patients with suspected or confirmed COVID-19 presenting with ACS. The proposed approach with ‘thrombolysis first’ for treating STEMI patients with suspected or confirmed COVID-19 infection could be unsafe in the case of underlying A-SCAD.
Collapse
Affiliation(s)
- Remo Albiero
- Interventional Cardiology Unit, Cardiovascular Department, Sondrio Hospital, Sondrio (SO), Italy
| | - Giuseppe Seresini
- Interventional Cardiology Unit, Cardiovascular Department, Sondrio Hospital, Sondrio (SO), Italy
| |
Collapse
|
40
|
Mandatori S, Pacella I, Marzolla V, Mammi C, Starace D, Padula F, Vitiello L, Armani A, Savoia C, Taurino M, De Zio D, Giampietri C, Piconese S, Cecconi F, Caprio M, Filippini A. Altered Tregs Differentiation and Impaired Autophagy Correlate to Atherosclerotic Disease. Front Immunol 2020; 11:350. [PMID: 32231663 PMCID: PMC7082762 DOI: 10.3389/fimmu.2020.00350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/13/2020] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is a progressive vascular disease representing the primary cause of morbidity and mortality in developed countries. Formerly, atherosclerosis was considered as a mere passive accumulation of lipids in blood vessels. However, it is now clear that atherosclerosis is a complex and multifactorial disease, in which the involvement of immune cells and inflammation play a key role. A variety of studies have shown that autophagy-a cellular catalytic mechanism able to remove injured cytoplasmic components in response to cellular stress-may be proatherogenic. So far, in this context, its role has been investigated in smooth muscle cells, macrophages, and endothelial cells, while the function of this catabolic protective process in lymphocyte functionality has been overlooked. The few studies carried out so far, however, suggested that autophagy modulation in lymphocyte subsets may be functionally related to plaque formation and development. Therefore, in this research, we aimed at better clarifying the role of lymphocyte subsets, mainly regulatory T cells (Tregs), in human atherosclerotic plaques and in animal models of atherosclerosis investigating the contribution of autophagy on immune cell homeostasis. Here, we investigate basal autophagy in a mouse model of atherosclerosis, apolipoprotein E (ApoE)-knockout (KO) mice, and we analyze the role of autophagy in driving Tregs polarization. We observed defective maturation of Tregs from ApoE-KO mice in response to tumor growth factor-β (TGFβ). TGFβ is a well-known autophagy inducer, and Tregs maturation defects in ApoE-KO mice seem to be related to autophagy impairment. In this work, we propose that autophagy underlies Tregs maturation, advocating that the study of this process in atherosclerosis may open new therapeutic strategies.
Collapse
Affiliation(s)
- Sara Mandatori
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Ilenia Pacella
- Laboratory of Cellular and Molecular Immunology, Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza Università di Roma, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Donatella Starace
- UOC, Clinical Pathology, San Giovanni Addolorata Hospital, Rome, Italy
| | - Fabrizio Padula
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Laura Vitiello
- Flow Cytometry Unit, IRCCS San Raffaele Pisana, Rome, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Carmine Savoia
- Cardiology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Taurino
- Unit of Vascular Surgery, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Daniela De Zio
- Cell Stress and Survival Unit, Center of Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Claudia Giampietri
- Unit of Human Anatomy, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Laboratory of Cellular and Molecular Immunology, Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza Università di Roma, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Center of Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Paediatric Haematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | - Antonio Filippini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
41
|
Lebedeva A, Maryukhnich E, Grivel JC, Vasilieva E, Margolis L, Shpektor A. Productive Cytomegalovirus Infection Is Associated With Impaired Endothelial Function in ST-Elevation Myocardial Infarction. Am J Med 2020; 133:133-142. [PMID: 31295440 PMCID: PMC6940528 DOI: 10.1016/j.amjmed.2019.06.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND An association between productive cytomegalovirus infection and atherosclerosis was shown recently in several trials, including a previous study of ours. However, the mechanism involved in this association is still under investigation. Here, we addressed the interaction between productive cytomegalovirus infection and endothelial function in patients with ST-elevation myocardial infarction (STEMI). METHODS We analyzed the presence of cytomegaloviral DNA in plasma and endothelial function in 33 patients with STEMI and 33 volunteers without cardiovascular diseases, using real-time polymerase chain reaction (PCR) and a noninvasive test of flow-mediated dilation. RESULTS Both the frequency of presence and the load of cytomegaloviral DNA were higher in plasma of patients with STEMI than those in controls. This difference was independent of other cardiovascular risk factors (7.38 [1.36-40.07]; P = 0.02). The results of the flow-mediated dilation test were lower in patients in STEMI than in controls (5.0% [2.65%-3.09%] vs 12. %5 [7.5%-15.15%]; P = 0.004) and correlated negatively with the cytomegaloviral DNA load (Spearman R = -0.407; P = 0.019) independently of other cardiovascular risk factors. CONCLUSIONS Productive cytomegalovirus infection in patients with STEMI correlated negatively with endothelial function independently of other cardiovascular risk factors. The impact of cytomegalovirus on endothelial function may explain the role of cytomegalovirus in cardiovascular prognosis.
Collapse
Affiliation(s)
- Anna Lebedeva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Elena Maryukhnich
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | | | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Md.
| | - Alexander Shpektor
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| |
Collapse
|
42
|
Kubátová H, Poledne R, Piťha J. Immune cells in carotid artery plaques: what can we learn from endarterectomy specimens? INT ANGIOL 2019; 39:37-49. [PMID: 31782285 DOI: 10.23736/s0392-9590.19.04250-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Endarterectomy specimens represent a unique opportunity to study atherosclerosis. This review aims to summarize the recent knowledge of atherogenesis from studies characterizing a cellular composition of carotid endarterectomy specimens. EVIDENCE ACQUISITION A non-systematic literature review was carried out to summarize recent knowledge regarding ex vivo analysis of carotid artery plaque composition. Upon evaluation of their relevance, and elaborate forward and backward search, 95 articles were included in the review. EVIDENCE SYNTHESIS Despite the significant advancement of in vivo imaging techniques, the stroke prediction based on carotid artery plaque morphology is not reliable. Besides analyses of plaque morphology, present studies focus on precise characterization of the different immune cell types and elucidation of their role in plaque development. Plaque content analyses revealed the presence of various immune cells in carotid artery plaques. Presence of different immune cells subpopulations can be connected to some undesirable changes in plaque stability. CONCLUSIONS Since the destabilization of the atherosclerotic plaque is a multifactorial process, a combination of various methods should be used to characterize the unstable plaques more accurately. In this context, studies characterizing plaque content from a cellular point of view could elucidate some processes underlying the plaque progression. Together with morphological evaluation, these analyses could enable more precise assessment of plaque stability.
Collapse
Affiliation(s)
- Hana Kubátová
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic - .,Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic -
| | - Rudolf Poledne
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Piťha
- Atherosclerosis Research Laboratory, Experimental Medicine Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.,Department of Internal Medicine, Second Medical Faculty, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
43
|
Schaftenaar FH, Amersfoort J, Douna H, Kröner MJ, Foks AC, Bot I, Slütter BA, van Puijvelde GHM, Drijfhout JW, Kuiper J. Induction of HLA-A2 restricted CD8 T cell responses against ApoB100 peptides does not affect atherosclerosis in a humanized mouse model. Sci Rep 2019; 9:17391. [PMID: 31757993 PMCID: PMC6874568 DOI: 10.1038/s41598-019-53642-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases form the most common cause of death worldwide, with atherosclerosis as main etiology. Atherosclerosis is marked by cholesterol rich lipoprotein deposition in the artery wall, evoking a pathogenic immune response. Characteristic for the disease is the pathogenic accumulation of macrophages in the atherosclerotic lesion, which become foam cells after ingestion of large quantities of lipoproteins. We hypothesized that, by inducing a CD8 T cell response towards lipoprotein derived apolipoprotein-B100 (ApoB100), lesional macrophages, that are likely to cross-present lipoprotein constituents, can specifically be eliminated. Based on in silico models for protein processing and MHC-I binding, 6 putative CD8 T cell epitopes derived from ApoB100 were synthesized. HLA-A2 binding was confirmed for all peptides by T2 cell binding assays and recall responses after vaccination with the peptides proved that 5 of 6 peptides could induce CD8 T cell responses. Induction of ApoB100 specific CD8 T cells did not impact plaque size and cellular composition in HLA-A2 and human ApoB100 transgenic LDLr−/− mice. No recall response could be detected in cultures of cells isolated from the aortic arch, which were observed in cell cultures of splenocytes and mesenteric lymph nodes, suggesting that the atherosclerotic environment impairs CD8 T cell activation.
Collapse
Affiliation(s)
- Frank H Schaftenaar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.
| | - Jacob Amersfoort
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Hidde Douna
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Mara J Kröner
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Bram A Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Gijs H M van Puijvelde
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Jan W Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.
| |
Collapse
|
44
|
Sieg SF, Bazdar DA, Zidar D, Freeman M, Lederman MM, Funderburg NT. Highly oxidized low-density lipoprotein mediates activation of monocytes but does not confer interleukin-1β secretion nor interleukin-15 transpresentation function. Immunology 2019; 159:221-230. [PMID: 31663113 DOI: 10.1111/imm.13142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/16/2019] [Accepted: 10/27/2019] [Indexed: 12/27/2022] Open
Abstract
Oxidized low-density lipoprotein (LDL) contributes to cardiovascular disease in part by mediating activation and maturation of monocytes and macrophages. Furthermore, co-localization studies using histochemical approaches have implicated a potential role for oxidized LDL as a mediator of interleukin-15 (IL-15) expression in myeloid cells of atherosclerotic plaque. The latter activity could be an important pro-inflammatory mechanism that mediates myeloid cell/T-cell crosstalk. Here, we examined the responses of primary human monocytes to highly oxidized LDL molecules. Oxidized LDL readily induced secretion of chemokines MCP-1 (CCL2) and GRO-α (CXCL1) but unlike lipopolysaccharide (LPS), has limited capacity to induce a variety of other cytokines including tumor necrosis factor-α, IL-6, IL-1β and interferon-γ-induced protein-10 and also displayed a poor capacity to induce p-Akt or P-S6 signaling. Failure of oxidized LDL to induce IL-1β secretion was associated with limited induction of caspase-1 activation. Furthermore, despite finding evidence that oxidized LDL could enhance the expression of IL-15 and IL-15 receptor expression in monocytes, we found no evidence that it could confer IL-15 transpresentation capability to these cells. This observation contrasted with induction of IL-15 transpresentation in lipopolysaccharide-stimulated monocytes. Overall, our data suggest that highly oxidized LDL is a selective inducer of monocyte activation. Sterile inflammatory mediators, particularly those implicated in Toll-like receptor 4 signaling, may play a role in vascular pathology but the activities of these agents are not uniform.
Collapse
Affiliation(s)
- Scott F Sieg
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Douglas A Bazdar
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David Zidar
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Michael Freeman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael M Lederman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, USA
| |
Collapse
|
45
|
Simons KH, de Vries MR, Peters HAB, Jukema JW, Quax PHA, Arens R. CD8+ T Cells Protect During Vein Graft Disease Development. Front Cardiovasc Med 2019; 6:77. [PMID: 31263704 PMCID: PMC6584838 DOI: 10.3389/fcvm.2019.00077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
Aims: Vein grafts are frequently used conduits for arterial reconstruction in patients with cardiovascular disease. Unfortunately, vein graft disease (VGD) causes diminished patency rates. Innate immune system components are known to contribute to VGD. However, the role of T cells has yet to be established. The purpose of this study was to investigate the role of T cells and T cell activation pathways via the T cell receptor (TCR), co-stimulation and bystander effect in VGD. Methods and results: Here, we show upon vein graft surgery in mice depleted of CD4+ T cells or CD8+ T cells, that CD8+ T cells are locally activated and have a major protective role for vein graft patency. In presence of CD8+ T cells vein grafts appear patent while CD8+ T cell depletion results in occluded vein grafts with increases apoptosis. Importantly, the protective effect of CD8+ T cells in VGD development was TCR and co-stimulation independent. This was demonstrated in vein grafts of OT-I mice, CD70−/−, CD80/86−/−, and CD70/80/86−/− mice compared to C57BL/6 mice. Interestingly, cytokines including IL-15, IL-18, IL-33, and TNF are up-regulated in vein grafts. These cytokines are co-operatively capable to activate CD8+ T cells in a bystander-mediated fashion, in contrast to CD4+ T cells. Conclusions: T cells are modulators of VGD with a specific protective role of CD8+ T cells, which are locally activated in vein grafts. CD8+ T cells may protect against occlusive lesions by providing survival signals, and concert their protection independent of TCR and co-stimulation signaling.
Collapse
Affiliation(s)
- Karin H Simons
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Margreet R de Vries
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Hendrika A B Peters
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
46
|
Neupane R, Jin X, Sasaki T, Li X, Murohara T, Cheng XW. Immune Disorder in Atherosclerotic Cardiovascular Disease - Clinical Implications of Using Circulating T-Cell Subsets as Biomarkers. Circ J 2019; 83:1431-1438. [PMID: 31092769 DOI: 10.1253/circj.cj-19-0114] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerotic cardiovascular disease (ACVD) is an inflammatory phenomenon that leads to structural abnormality in the vascular lumen due to the formation of atheroma by the deposition of lipid particles and inflammatory cytokines. There is a close interaction between innate immune cells (neutrophils, monocyte, macrophages, dendritic cells) and adaptive immune cells (T and B lymphocytes) in the initiation and progression of atherosclerosis. According to novel insights into the role of adaptive immunity in atherosclerosis, the activation of CD4+T cells in response to oxidized low-density lipoprotein-antigen initiates the formation and facilitates the propagation of atheroma, whereas CD8+T cells cause the rupture of a developed atheroma by their cytotoxic nature. Peripheral CD4+and CD8+T-cell counts were altered in patients with other cardiovascular risk factors. Furthermore, on evaluation of the feasibility of immune cells as a diagnostic tool, the blood CD4+(helper), CD8+(cytotoxic), and CD4+CD25+Foxp3+(regulatory) T cells and the ratio of CD4 to CD8 cells hold promise as biomarkers of coronary artery disease and their subtypes. T cells also could be a therapeutic target for cardiovascular diseases. The goal of this review was therefore to summarize the available information regarding immune disorders in ACVD with a special focus on the clinical implications of circulating T-cell subsets as biomarkers.
Collapse
Affiliation(s)
- Rajib Neupane
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Xiongjie Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine
| | - Xiang Li
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital.,Department of Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|
47
|
Lebedeva AM, Shpektor AV, Vasilieva EY, Margolis LB. Cytomegalovirus Infection in Cardiovascular Diseases. BIOCHEMISTRY (MOSCOW) 2019; 83:1437-1447. [PMID: 30878019 DOI: 10.1134/s0006297918120027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Atherosclerosis underlies the development of many cardiovascular diseases that continue to hold a leading place among the causes of death in developed countries. The role of activated immune cells in atherosclerosis progression has been convincingly demonstrated, but the mechanism of their action remains poorly investigated. Since atherosclerosis is associated with chronic inflammatory response, involvement of viral and bacterial infections in atherogenesis has been examined. A special place among the infectious agents is held by human herpesviruses as the most common persistent viruses in human population coupled to chronic inflammation during atherosclerosis. We found that activation of cytomegalovirus (CMV, human herpesvirus 5) infection is associated with the emergence of acute coronary syndrome, which is in a good agreement with the data on productive CMV infection published elsewhere. In this review, we discuss the data obtained by us and other researchers regarding the role of cytomegalovirus infection and related potential mechanisms resulting in the expansion of atherosclerotic plaques during ischemic heart disease and stroke, including virus transfer to immune and endothelial cells via extracellular vesicles. In particular, the data presented in the review demonstrate that virus spreading in the vascular wall triggers immune system activation in atherosclerotic plaques and causes endothelial dysfunction. Moreover, productive CMV infection in patients with acute myocardial infarction correlates with the extent of endothelial dysfunction. The mechanisms described by us and other researchers may explain the role of CMV infection in atherosclerosis and development of ischemic heart disease.
Collapse
Affiliation(s)
- A M Lebedeva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia.
| | - A V Shpektor
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - E Yu Vasilieva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - L B Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
Kritikou E, van der Heijden T, Swart M, van Duijn J, Slütter B, Wezel A, Smeets HJ, Maffia P, Kuiper J, Bot I. Hypercholesterolemia Induces a Mast Cell-CD4 + T Cell Interaction in Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 202:1531-1539. [PMID: 30683705 DOI: 10.4049/jimmunol.1800648] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/20/2018] [Indexed: 11/19/2022]
Abstract
Mast cells (MCs) are potent innate immune cells that aggravate atherosclerosis through the release of proinflammatory mediators inside atherosclerotic plaques. Similarly, CD4+ T cells are constituents of the adaptive immune response and accumulate within the plaques following lipid-specific activation by APCs. Recently it has been proposed that these two cell types can interact in a direct manner. However, no indication of such an interaction has been investigated in the context of atherosclerosis. In our study, we aimed to examine whether MCs can act as APCs in atherosclerosis, thereby modulating CD4+ T cell responses. We observed that MCs increased their MHC class II expression under hyperlipidemic conditions both in vivo and in vitro. Furthermore, we showed that MCs can present Ags in vivo via MHC class II molecules. Serum from high-fat diet-fed mice also enhanced the expression of the costimulatory molecule CD86 on cultured MCs, whereas OVA peptide-loaded MCs increased OT-II CD4+ T cell proliferation in vitro. The aortic CD4+ and TH1 cell content of atherosclerotic mice that lack MCs was reduced as compared with their wild-type counterparts. Importantly, we identified MCs that express HLA-DR in advanced human atheromata, indicating that these cells are capable of Ag presentation within human atherosclerotic plaques. Therefore, in this artice, we show that MCs may directly modulate adaptive immunity by acting as APCs in atherosclerosis.
Collapse
Affiliation(s)
- Eva Kritikou
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands;
| | - Thomas van der Heijden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Maarten Swart
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom; and.,Department of Pharmacy, University of Naples Federico II, 80138 Naples, Italy
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| |
Collapse
|
49
|
Loguinova M, Pinegina N, Kogan V, Vagida M, Arakelyan A, Shpektor A, Margolis L, Vasilieva E. Monocytes of Different Subsets in Complexes with Platelets in Patients with Myocardial Infarction. Thromb Haemost 2018; 118:1969-1981. [PMID: 30300910 DOI: 10.1055/s-0038-1673342] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Acute myocardial infarction (AMI) is associated with activation of various cells, including platelets that form monocyte-platelet complexes (MPCs). Here, we analysed MPC in vivo and in vitro and investigated the abilities of different monocyte subclasses to form MPC, the characteristics of the cells involved in MPC formation and MPC changes in AMI. We identified MPC by co-staining for platelet antigen CD41a and monocyte antigens CD14 and CD16. Platelet activation was evaluated from expression of phosphatidylserine as revealed by annexin V. Our results confirm published data and provide new information regarding the patterns of MPC in AMI patients. We found that the patterns of platelet aggregation with monocytes were different in AMI patients and controls: (1) in AMI patients, MPC formed by intermediate monocytes carry more platelets whereas in healthy controls more platelets aggregated with classical monocytes; (2) the numbers of MPC in AMI patients, being already higher than in controls, were further increased if these patients suffered various in-hospital complications; (3) on the basis of the CD41a fluorescence of the antibody-stained MPC, some of the aggregates seem to consist of monocytes and platelet-derived extracellular vesicles (EVs); (4) aggregation of monocytes with platelet EV occurred in in vitro experiments; and (5) these experiments demonstrated that monocytes from AMI patients aggregate with both platelets and platelet EVs more efficiently than do monocytes from controls. MPC in AMI patients may play an important role in this pathology.
Collapse
Affiliation(s)
- Marina Loguinova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Natalia Pinegina
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Valeria Kogan
- Ariel University, Institute for Translational Research, Ariel, Israel
| | - Murad Vagida
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Anush Arakelyan
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States
| | - Alexander Shpektor
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| |
Collapse
|
50
|
Lacy M, Atzler D, Liu R, de Winther M, Weber C, Lutgens E. Interactions between dyslipidemia and the immune system and their relevance as putative therapeutic targets in atherosclerosis. Pharmacol Ther 2018; 193:50-62. [PMID: 30149100 DOI: 10.1016/j.pharmthera.2018.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) continues to be a leading cause of death worldwide with atherosclerosis being the major underlying pathology. The interplay between lipids and immune cells is believed to be a driving force in the chronic inflammation of the arterial wall during atherogenesis. Atherosclerosis is initiated as lipid particles accumulate and become trapped in vessel walls. The subsequent immune response, involving both adaptive and immune cells, progresses plaque development, which may be exacerbated under dyslipidemic conditions. Broad evidence, especially from animal models, clearly demonstrates the effect of lipids on immune cells from their development in the bone marrow to their phenotypic switching in circulation. Interestingly, recent research has also shown a long-lasting epigenetic signature from lipids on immune cells. Traditionally, cardiovascular therapies have approached atherosclerosis through lipid-lowering medications because, until recently, anti-inflammatory therapies have been largely unsuccessful in clinical trials. However, the recent Canakinumab Antiinflammatory Thrombosis Outcomes Study (CANTOS) provided pivotal support of the inflammatory hypothesis of atherosclerosis in man spurring on anti-inflammatory strategies to treat atherosclerosis. In this review, we describe the interactions between lipids and immune cells along with their specific outcomes as well as discuss their future perspective as potential cardiovascular targets.
Collapse
Affiliation(s)
- Michael Lacy
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Rongqi Liu
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Menno de Winther
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|