1
|
Natarajan N, Dutta P. 'Training' of innate immunity following myocardial infarction exacerbates atherosclerosis. Eur Heart J 2024; 45:685-687. [PMID: 38271239 PMCID: PMC11491281 DOI: 10.1093/eurheartj/ehae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Affiliation(s)
- Niranjana Natarajan
- Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Partha Dutta
- Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
2
|
Dong Z, Hou L, Luo W, Pan LH, Li X, Tan HP, Wu RD, Lu H, Yao K, Mu MD, Gao CS, Weng XY, Ge JB. Myocardial infarction drives trained immunity of monocytes, accelerating atherosclerosis. Eur Heart J 2024; 45:669-684. [PMID: 38085922 DOI: 10.1093/eurheartj/ehad787] [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: 11/03/2022] [Revised: 09/28/2023] [Accepted: 11/16/2023] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND AND AIMS Survivors of acute coronary syndromes face an elevated risk of recurrent atherosclerosis-related vascular events despite advanced medical treatments. The underlying causes remain unclear. This study aims to investigate whether myocardial infarction (MI)-induced trained immunity in monocytes could sustain proatherogenic traits and expedite atherosclerosis. METHODS Apolipoprotein-E deficient (ApoE-/-) mice and adoptive bone marrow transfer chimeric mice underwent MI or myocardial ischaemia-reperfusion (IR). A subsequent 12-week high-fat diet (HFD) regimen was implemented to elucidate the mechanism behind monocyte trained immunity. In addition, classical monocytes were analysed by flow cytometry in the blood of enrolled patients. RESULTS In MI and IR mice, blood monocytes and bone marrow-derived macrophages exhibited elevated spleen tyrosine kinase (SYK), lysine methyltransferase 5A (KMT5A), and CCHC-type zinc finger nucleic acid-binding protein (CNBP) expression upon exposure to a HFD or oxidized LDL (oxLDL) stimulation. MI-induced trained immunity was transmissible by transplantation of bone marrow to accelerate atherosclerosis in naive recipients. KMT5A specifically recruited monomethylation of Lys20 of histone H4 (H4K20me) to the gene body of SYK and synergistically transactivated SYK with CNBP. In vivo small interfering RNA (siRNA) inhibition of KMT5A or CNBP potentially slowed post-MI atherosclerosis. Sympathetic denervation with 6-hydroxydopamine reduced atherosclerosis and inflammation after MI. Classical monocytes from ST-elevation MI (STEMI) patients with advanced coronary lesions expressed higher SYK and KMT5A gene levels. CONCLUSIONS The findings underscore the crucial role of monocyte trained immunity in accelerated atherosclerosis after MI, implying that SYK in blood classical monocytes may serve as a predictive factor for the progression of atherosclerosis in STEMI patients.
Collapse
Affiliation(s)
- Zheng Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lei Hou
- Institute of Cardiovascular Diseases, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, China
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai 201600, China
| | - Wei Luo
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Li-Hong Pan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiao Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hai-Peng Tan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Run-Da Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Kang Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Man-Di Mu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chen-Shan Gao
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Xin-Yu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jun-Bo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Zeng GG, Zhou J, Jiang WL, Yu J, Nie GY, Li J, Zhang SQ, Tang CK. A Potential Role of NFIL3 in Atherosclerosis. Curr Probl Cardiol 2024; 49:102096. [PMID: 37741601 DOI: 10.1016/j.cpcardiol.2023.102096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Nuclear factor interleukin-3 (NFIL3), a proline- and acidic-residue-rich (PAR) bZIP transcription factor, is called the E4 binding protein 4 (E4BP4) as well, which is relevant to regulate the circadian rhythms and the viability of cells. More and more evidence has shown that NFIL3 is associated with different cardiovascular diseases. In recent years, it has been found that NFIL3 has significant functions in the progression of atherosclerosis (AS) via the regulation of inflammatory response, macrophage polarization, some immune cells and lipid metabolism. In this overview, we sum up the function of NFIL3 during the development of AS and offer meaningful views how to treat cardiovascular disease related to AS.
Collapse
Affiliation(s)
- Guang-Gui Zeng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jing Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; School of Pharmaceutical Science, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jiang Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Gui-Ying Nie
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2019 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jing Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Shi-Qian Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
4
|
Ding J, Li H, Liu W, Wang X, Feng Y, Guan H, Chen Z. miR-186-5p Dysregulation in Serum Exosomes from Patients with AMI Aggravates Atherosclerosis via Targeting LOX-1. Int J Nanomedicine 2022; 17:6301-6316. [PMID: 36536941 PMCID: PMC9758944 DOI: 10.2147/ijn.s383904] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose The formation of macrophage-derived foam cells via the uptake of modified lipoproteins is a pivotal development event in atherosclerosis. It has been reported that clinical and experimental myocardial infarction could accelerate atherosclerosis. Several studies have suggested the critical role of exosomes in cardiovascular diseases. However, the role of exosomes from patients with acute myocardial infarction (AMI) patients in atherogenesis remains unclear. Patients and Methods Serum exosomes from AMI patients (AMI-Exo) and control individuals (Con-Exo) were isolated and characterized. These exosomes were studied in vitro and in vivo to determine their impact on macrophage foaming and atherogenesis. Results Our results showed that AMI-Exo promoted foam cell formation in oxidized low-density lipoprotein (ox-LDL)-treated macrophages and progression of atherosclerosis in high-fat/cholesterol diet-fed ApoE-/- mice together with a significantly upregulated levels of lectin-like ox-LDL receptor-1 (LOX-1). The miR-186-5p was found to be downregulated in AMI-Exo and macrophages administered with AMI-Exo. Moreover, serum exosomal miR-186-5p achieved high diagnostic performance for AMI. Luciferase reporter assay indicated that miR-186-5p directly inhibited LOX-1. The endogenous or exogenous miR-186-5p deficiency enhanced lipid accumulation by upregulating LOX-1, whereas miR-186-5p mimics had a reverse effect. Conclusion In conclusion, the current findings suggest that dysregulated miR-186-5p in AMI-Exo may explain the contribution of acute ischemia events to the advancement of atherosclerosis by enhancing macrophage foaming via its target, LOX-1.
Collapse
Affiliation(s)
- Jiaxing Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Henan Provincial People’s Hospital Heart Center, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Huili Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Wei Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xuehua Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu Feng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hongquan Guan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Correspondence: Zhijian Chen; Hongquan Guan, Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China, Tel + 86 27 85726011, Fax +86 27 85727340, Email
| | - Zhijian Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
5
|
Kyaw T, Loveland P, Kanellakis P, Cao A, Kallies A, Huang AL, Peter K, Toh BH, Bobik A. Alarmin-activated B cells accelerate murine atherosclerosis after myocardial infarction via plasma cell-immunoglobulin-dependent mechanisms. Eur Heart J 2021; 42:938-947. [PMID: 33338208 DOI: 10.1093/eurheartj/ehaa995] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/30/2020] [Accepted: 11/29/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Paula Loveland
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, University of Melbourne, 792 Elizabeth Street, Melbourne, Vic 3000, Australia
| | - Alex L Huang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
6
|
Chen Y, Zhang H, Hu L, Shi H, Liu X, Jia J, Sun S, Ou Y, Luo X, Zhou G, Shen W. Pravastatin attenuates atherosclerosis after myocardial infarction by inhibiting inflammatory Ly6C high monocytosis in apolipoprotein E knockout mice. J Int Med Res 2021; 48:300060520932816. [PMID: 32662710 PMCID: PMC7361503 DOI: 10.1177/0300060520932816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Objective To evaluate the protective effect of pravastatin on atherosclerotic development and inflammatory monocyte subset in atherosclerotic apolipoprotein E (ApoE)−/− mice after myocardial infarction (MI). Methods Male ApoE−/− mice (8 weeks old) were fed a high-fat diet for 14 weeks throughout the experiment. A MI model was produced using 18-week-old ApoE−/− mice. They were randomly divided into three groups: sham group, MI group, and MI+Pra group (40 mg/kg/day pravastatin). After 4 weeks (at the end of the study period), the mice were sacrificed and cardiac function was evaluated by echocardiography. Aortic lesion areas were evaluated using oil red O staining. Plaque macrophage in aortic sinus was analyzed by immunofluorescence staining. Flow cytometry was used to explore the proportions of monocyte subsets in the blood, spleen, and bone marrow. Results Pravastatin improved cardiac function and reduced lesion areas. It also attenuated the supply of monocytes in spleen, especially the inflammatory Ly6Chigh monocyte subset. Pravastatin also subsequently reduced macrophage accumulation in atherosclerotic lesions. Conclusions MI accelerated chronic atherosclerosis progress. Pravastatin suppressed atherosclerotic development and inhibited inflammatory monocytosis after MI in ApoE−/− mice.
Collapse
Affiliation(s)
- Yufei Chen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongqi Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaojin Liu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianguo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shengjia Sun
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Ou
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinping Luo
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guomin Zhou
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Shen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Beijnink CWH, Thim T, van der Heijden DJ, Klem I, Al-Lamee R, Vos JL, Koop Y, Dijkgraaf MGW, Beijk MAM, Kim RJ, Davies J, Raposo L, Baptista SB, Escaned J, Piek JJ, Maeng M, van Royen N, Nijveldt R. Instantaneous wave-free ratio guided multivessel revascularisation during percutaneous coronary intervention for acute myocardial infarction: study protocol of the randomised controlled iMODERN trial. BMJ Open 2021; 11:e044035. [PMID: 33452200 PMCID: PMC7813313 DOI: 10.1136/bmjopen-2020-044035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Recent randomised clinical trials showed benefit of non-culprit lesion revascularisation in ST-elevation myocardial infarction (STEMI) patients. However, it remains unclear whether revascularisation should be performed at the index procedure or at a later stage. METHODS AND ANALYSIS The instantaneous wave-free ratio (iFR) Guided Multivessel Revascularisation During Percutaneous Coronary Intervention for Acute Myocardial Infarction trial is a multicentre, randomised controlled prospective open-label trial with blinded evaluation of endpoints. After successful primary percutaneous coronary intervention (PCI), eligible STEMI patients with residual non-culprit lesions are randomised, to instantaneous wave-free ratio guided treatment of non-culprit lesions during the index procedure versus deferred cardiac MR-guided management within 4 days to 6 weeks. The primary endpoint of the study is the combined occurrence of all-cause death, recurrent myocardial infarction and hospitalisation for heart failure at 12 months follow-up. Clinical follow-up includes questionnaires at 3 months and outpatient visits at 6 months and 12 months after primary PCI. Furthermore, a cost-effectiveness analysis will be performed. ETHICS AND DISSEMINATION Permission to conduct this trial has been granted by the Medical Ethical Committee of the Amsterdam University Medical Centres (loc. VUmc, ID NL60107.029.16). The primary results of this trial will be shared in a main article and subgroup analyses or spin-off studies will be shared in secondary papers. TRIAL REGISTRATION NUMBER NCT03298659.
Collapse
Affiliation(s)
| | - Troels Thim
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | | | - Igor Klem
- Cardiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Rasha Al-Lamee
- Cardiology, Imperial College London Faculty of Medicine, London, UK
| | | | - Yvonne Koop
- Cardiology, Radboudumc, Nijmegen, The Netherlands
| | - Marcel G W Dijkgraaf
- Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC - Locatie AMC, Amsterdam, The Netherlands
| | - Marcel A M Beijk
- Cardiology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Raymond J Kim
- Cardiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Justin Davies
- Faculty of Medicine, Imperial College London, London, UK
| | - Luis Raposo
- Cardiology, Centro Hospitalar de Lisboa Ocidental EPE, Lisboa, Portugal
| | | | - Javier Escaned
- Interventional Cardiology, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Jan J Piek
- Cardiology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Michael Maeng
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | | | | |
Collapse
|
8
|
Güner A, Zehİr R, KalçIk M, Uslu A, Ösken A, Kalkan AK, Güner EG. Eosinophil percentage as a new prognostic marker in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Interv Med Appl Sci 2020; 11:146-153. [PMCID: PMC9467337 DOI: 10.1556/1646.11.2019.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 11/19/2022] Open
Abstract
Background In addition to proinflammatory properties, eosinophils can stimulate platelet activation and enhance prothrombotic pathways. In this study, we aimed to investigate the association between the eosinophil percentage (EOS%) and major adverse cardiac events (MACE) in patients with ST-segment elevation myocardial infarction (STEMI). Methods This study enrolled a total of 1,909 patients who were diagnosed with STEMI. Ventricular arrhythmia, reinfarction, the need for cardiopulmonary resuscitation, target vessel revascularization, congestive heart failure, and cardiovascular mortality during index hospitalization were defined as MACE. Results Three hundred and eighty patients (19.7%) reached the combined endpoint with MACE. The rates of inhospital mortality and MACE were significantly higher in low EOS% group as compared to high EOS% group (4% vs. 1.1%, p < 0.01 and 32.8% vs. 11.3%, p < 0.01, respectively). On multivariate logistic regression analyses, EOS% (OR = 0.44, p < 0.01) was found to be one of the independent predictors of MACE. The EOS% lower than 0.60 on admission predicted inhospital MACE with a sensitivity of 68% and a specificity of 72% (AUC: 0.684, p < 0.01). Conclusions Low EOS% on admission may be associated with high inhospital MACE in STEMI patients. EOS% may be used as a novel biomarker for risk stratification of these patients.
Collapse
Affiliation(s)
- Ahmet Güner
- 1 Department of Cardiology, Kartal Kosuyolu Training and Research Hospital, Istanbul, Turkey
| | - Regayİp Zehİr
- 2 Department of Cardiology, Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Macİt KalçIk
- 3 Department of Cardiology, Faculty of Medicine, Hitit University, Çorum, Turkey
| | - Abdulkadİr Uslu
- 1 Department of Cardiology, Kartal Kosuyolu Training and Research Hospital, Istanbul, Turkey
| | - Altuğ Ösken
- 2 Department of Cardiology, Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Alİ Kemal Kalkan
- 4 Department of Cardiology, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ezgİ Gültekİn Güner
- 4 Department of Cardiology, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
9
|
Abstract
A central feature of atherosclerosis, the most prevalent chronic vascular disease and root cause of myocardial infarction and stroke, is leukocyte accumulation in the arterial wall. These crucial immune cells are produced in specialized niches in the bone marrow, where a complex cell network orchestrates their production and release. A growing body of clinical studies has documented a correlation between leukocyte numbers and cardiovascular disease risk. Understanding how leukocytes are produced and how they contribute to atherosclerosis and its complications is, therefore, critical to understanding and treating the disease. In this review, we focus on the key cells and products that regulate hematopoiesis under homeostatic conditions, during atherosclerosis and after myocardial infarction.
Collapse
Affiliation(s)
- Wolfram C Poller
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Matthias Nahrendorf
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Radiology (M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Filip K Swirski
- From the Center for Systems Biology (W.C.P., M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Radiology (M.N., F.K.S.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
10
|
Abstract
Atherosclerotic cardiovascular disease is a leading cause of death and morbidity globally. Over the past several years, arterial inflammation has been implicated in the pathophysiology of athero-thrombosis, substantially confirming what pathologist Rudolf Virchow had observed in the 19th century. Lipid lowering, lifestyle changes, and modification of other risk factors have reduced cardiovascular complications of athero-thrombosis, but a substantial residual risk remains. In view of the pathogenic role of inflammation in athero-thrombosis, directly targeting inflammation has emerged as an additional potential therapeutic option; and some early promising results have been suggested by the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS), in which canakinumab, a fully human monoclonal antibody targeting the pro-inflammatory and pro-atherogenic cytokine interleukin 1 beta, was shown to reduce cardiovascular events.
Collapse
Affiliation(s)
- Prediman K Shah
- Helga and Walter Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Dalgisio Lecis
- Helga and Walter Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Cardiovascular Medicine, "Tor Vergata" University of Rome, Rome, Italy
| |
Collapse
|
11
|
Angiography-guided Multivessel Percutaneous Coronary Intervention Versus Ischemia-guided Percutaneous Coronary Intervention Versus Medical Therapy in the Management of Significant Disease in Non-Infarct-related Arteries in ST-Elevation Myocardial Infarction Patients With Multivessel Coronary Disease. Crit Pathw Cardiol 2019; 17:77-82. [PMID: 29768315 DOI: 10.1097/hpc.0000000000000144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND In ST-elevation myocardial infarction (STEMI) patients with multivessel (MV) disease, after primary percutaneous coronary intervention (PCI), emerging evidence suggests that significant disease in non-infarct-related coronary arteries (IRAs) should be routinely stented. Whether this procedure should be guided by angiography alone or ischemia testing is unclear. METHODS All STEMI patients treated with primary PCI between January 1, 2005, and December 31, 2012, at a tertiary cardiology center were reviewed retrospectively. Inclusion criterion is patients with at least 70% stenosis in non-IRAs. There were 3 treatment groups: (1) angiography-guided MV-PCI, (2) ischemia-guided PCI, and (3) medical therapy. Primary endpoint is all-cause mortality, and secondary end point is major adverse cardiovascular events (MACE), including death, acute coronary syndrome, revascularization, or stent thrombosis. Event-free survivals were compared using multivariate Cox proportional-hazards analysis. A propensity score-adjusted analysis was performed. RESULTS Four hundred forty-seven STEMI patients had >70% stenosis in non-IRAs. For all-cause mortality, the 3 strategies did not differ. For MACE, ischemia-guided PCI was associated with the lowest MACE rate, followed by angiography-guided PCI and medical therapy, which was associated with the highest MACE rate, driven by death and myocardial infarction. Hazard ratios (HRs) for MACE: angiography-guided MV-PCI versus ischemia-guided MV-PCI: HR = 2.23 [95% confidence interval (CI), 1.11-4.48; P = 0.023]; medical therapy versus angiography-guided MV-PCI: HR = 1.58 (95% CI, 0.99-2.63; P = 0.062); medical therapy versus ischemia-guided MV-PCI: HR = 1.72 (95% CI, 1.08-2.74; P = 0.022). Propensity score-adjusted analysis yielded similar results. CONCLUSIONS After primary PCI, complete revascularization in STEMI multivessel disease is associated with lower MACE rates than medical therapy. However, ischemia-testing-guided rather than angiography-guided revascularization was associated with the lowest MACE. This study provides preliminary data and hypotheses for future randomized controlled studies.
Collapse
|
12
|
Shah PK. Inflammation, infection and atherosclerosis. Trends Cardiovasc Med 2019; 29:468-472. [PMID: 30733074 DOI: 10.1016/j.tcm.2019.01.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/14/2019] [Accepted: 01/20/2019] [Indexed: 12/12/2022]
Abstract
Atherosclerotic cardiovascular disease is a leading cause of death in much of the world. Adoption of a healthy lifestyle and cholesterol lowering are the key measures used to prevent major complications of atherosclerosis. Recent data have identified a critical role for inflammation mediated through activation of both innate and adaptive immune pathways in the pathophysiology of atherosclerosis opening up opportunities for development of anti-inflammatory interventions that could supplement risk factor modification and lipid lowering as an approach to further reducing the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Prediman K Shah
- The Helga and Walter Oppenheimer Atherosclerosis Research Center, Cardiology Division and Smidt Heart Institute at Cedars Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States.
| |
Collapse
|
13
|
Taneja G, Sud A, Pendse N, Panigrahi B, Kumar A, Sharma AK. Nano-medicine and Vascular Endothelial Dysfunction: Options and Delivery Strategies. Cardiovasc Toxicol 2018; 19:1-12. [DOI: 10.1007/s12012-018-9491-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Twelve-month clinical outcomes of acute non-ST versus ST-segment elevation myocardial infarction patients with reduced preprocedural thrombolysis in myocardial infarction flow undergoing percutaneous coronary intervention. Coron Artery Dis 2018; 29:416-422. [PMID: 29708887 DOI: 10.1097/mca.0000000000000632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Reduced preprocedural thrombolysis in myocardial infarction (TIMI) flow in patients with ST-segment elevation myocardial infarction (STEMI) is known to be associated with increased mortality. However, clinical implications of reduced preprocedural TIMI flow in patients with non-ST-segment elevation myocardial infarction (NSTEMI) have not been fully elucidated as yet. The aim of the present study was to compare the clinical influence of reduced preprocedural TIMI flows between patients with STEMI and NSTEMI undergoing percutaneous coronary intervention (PCI). METHODS From the Korea Acute Myocardial Infarction Registry, a total of 7336 AMI patients with angiographically confirmed reduced preprocedural TIMI flow (TIMI 0/1) during PCI were selected and divided into STEMI (n=4852) and NSTEMI (n=2484) groups. The 12-month composite of total death, nonfatal myocardial infarction, coronary artery bypass graft, and repeated PCI was compared between the two groups. RESULTS After adjustment of baseline confounders by propensity score stratification, the NSTEMI group had lower incidences of major adverse cardiac events than the STEMI group (7.15 vs. 11.19%; hazard ratio: 0.63; 95% confidence interval: 0.47-0.84; P=0.001) at 12 months, which was largely attributable to the lower incidences of total deaths (2.43 vs. 3.99%; P=0.04) and repeated PCI (3.81 vs. 6.41%; P=0.01). CONCLUSION Among AMI patients with TIMI 0/1, patients with NSTEMI had better outcomes compared with those of patients with STEMI on the basis of the incidences of 12-month outcomes. This could be attributable to lower total death and repeated revascularization in patients with NSTEMI.
Collapse
|
15
|
Postnatal undernutrition in mice causes cardiac arrhythmogenesis which is exacerbated when pharmacologically stressed. J Dev Orig Health Dis 2018; 9:417-424. [DOI: 10.1017/s2040174418000156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AbstractGrowth restriction caused by postnatal undernutrition increases risk for cardiovascular disease in adulthood with the potential to induce arrhythmogenesis. Thus, the purpose was to determine if undernutrition during development produced arrhythmias at rest and when stressed with dobutamine in adulthood. Mouse dams were fed (CON: 20% protein), or low-protein (LP: 8%) diet before mating. A cross-fostering model was used where pups nursed by dams fed LP diet in early [EUN; postnatal day (PN) 1–10], late (LUN; PN11–21) and whole (PUN; 1–21) phases of postnatal life. Weaned pups were switched to CON diets for the remainder of the study (PN80). At PN80, body composition (magnetic resonance imaging), and quantitative electrocardiogram (ECG) measurements were obtained under 1% isoflurane anesthesia. After baseline ECG, an IP injection (1.5 µg/g body weight) of dobutamine was administered and ECG repeated. Undernutrition significantly (P<0.05) reduced body weight in LUN (22.68±0.88 g) and PUN (19.96±0.32 g) but not in CON (25.05±0.96 g) and EUN (25.28±0.9207 g). Fat mass decreased in all groups compared with controls (CON: 8.00±1.2 g, EUN: 6.32±0.65 g, LUN: 5.11±1.1 g, PUN: 3.90±0.25 g). Lean mass was only significantly reduced in PUN (CON: 17.99±0.26 g, EUN: 17.78±0.39 g, LUN: 17.34±0.33 g, PUN: 15.85±0.28 g). Absolute heart weights were significantly less from CON, with PUN having the smallest. ECG showed LUN had occurrences of atrial fibrillation; EUN had increases of 1st degree atrioventricular block upon stimulation, and PUN had increased risk for ventricular depolarization arrhythmias. CON did not display arrhythmias. Undernutrition in early life resulted in ventricular arrhythmias under stressed conditions, but undernutrition occurring in later postnatal life there is an increased incidence of atrial arrhythmias.
Collapse
|
16
|
Zhou H, Ma Q, Zhu P, Ren J, Reiter RJ, Chen Y. Protective role of melatonin in cardiac ischemia-reperfusion injury: From pathogenesis to targeted therapy. J Pineal Res 2018; 64. [PMID: 29363153 DOI: 10.1111/jpi.12471] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023]
Abstract
Acute myocardial infarction (MI) is a major cause of mortality and disability worldwide. In patients with MI, the treatment option for reducing acute myocardial ischemic injury and limiting MI size is timely and effective myocardial reperfusion using either thombolytic therapy or primary percutaneous coronary intervention (PCI). However, the procedure of reperfusion itself induces cardiomyocyte death, known as myocardial reperfusion injury, for which there is still no effective therapy. Recent evidence has depicted a promising role of melatonin, which possesses powerful antioxidative and anti-inflammatory properties, in the prevention of ischemia-reperfusion (IR) injury and the protection against cardiomyocyte death. A number of reports explored the mechanism of action behind melatonin-induced beneficial effects against myocardial IR injury. In this review, we summarize the research progress related to IR injury and discuss the unique actions of melatonin as a protective agent. Furthermore, the possible mechanisms responsible for the myocardial benefits of melatonin against reperfusion injury are listed with the prospect of the use of melatonin in clinical application.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Zhou H, Li D, Zhu P, Hu S, Hu N, Ma S, Zhang Y, Han T, Ren J, Cao F, Chen Y. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARγ/FUNDC1/mitophagy pathways. J Pineal Res 2017; 63. [PMID: 28749565 DOI: 10.1111/jpi.12438] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/24/2017] [Indexed: 12/16/2022]
Abstract
Platelet activation is a major (patho-) physiological mechanism that underlies ischemia/reperfusion (I/R) injury. In this study, we explored the molecular signals for platelet hyperactivity and investigated the beneficial effects of melatonin on platelet reactivity in response to I/R injury. After reperfusion, peroxisome proliferator-activated receptor γ (PPARγ) was progressively downregulated in patients with acute myocardial infarction undergoing coronary artery bypass grafting (CABG) surgery and in mice with I/R injury model. Loss of PPARγ was closely associated with FUN14 domain containing 1 (FUNDC1) dephosphorylation and mitophagy activation, leading to increased mitochondrial electron transport chain complex (ETC.) activity, enhanced mitochondrial respiratory function, and elevated ATP production. The improved mitochondrial function strongly contributed to platelet aggregation, spreading, expression of P-selectin, and final formation of micro-thromboses, eventually resulting in myocardial dysfunction and microvascular structural destruction. However, melatonin powerfully suppressed platelet activation via restoration of the PPARγ content in platelets, which subsequently blocked FUNDC1-required mitophagy, mitochondrial energy production, platelet hyperactivity, and cardiac I/R injury. In contrast, genetic ablation of PPARγ in platelet abolished the beneficial effects of melatonin on mitophagy, mitochondrial ATP supply, and platelet activation. Our results lay the foundation for the molecular mechanism of platelet activation in response to I/R injury and highlight that the manipulation of the PPARγ/FUNDC1/mitophagy pathway by melatonin could be a novel strategy for cardioprotection in the setting of cardiac I/R injury.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Dandan Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Nan Hu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Sai Ma
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Tianwen Han
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW With the intention to summarize the currently available evidence on the pathophysiological relevance of inflammation in heart failure, this review addresses the question whether inflammation is a cause or consequence of heart failure, or both. RECENT FINDINGS This review discusses the diversity (sterile, para-inflammation, chronic inflammation) and sources of inflammation and gives an overview of how inflammation (local versus systemic) can trigger heart failure. On the other hand, the review is outlined how heart failure-associated wall stress and signals released by stressed, malfunctioning, or dead cells (DAMPs: e.g., mitochondrial DNA, ATP, S100A8, matricellular proteins) induce cardiac sterile inflammation and how heart failure provokes inflammation in various peripheral tissues in a direct (inflammatory) and indirect (hemodynamic) manner. The crosstalk between the heart and peripheral organs (bone marrow, spleen, gut, adipose tissue) is outlined and the importance of neurohormonal mechanisms including the renin angiotensin aldosteron system and the ß-adrenergic nervous system in inflammation and heart failure is discussed. Inflammation and heart failure are strongly interconnected and mutually reinforce each other. This indicates the difficulty to counteract inflammation and heart failure once this chronic vicious circle has started and points out the need to control the inflammatory process at an early stage avoiding chronic inflammation and heart failure. The diversity of inflammation further addresses the need for a tailored characterization of inflammation enabling differentiation of inflammation and subsequent target-specific strategies. It is expected that the characterization of the systemic and/or cardiac immune profile will be part of precision medicine in the future of cardiology.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Tschöpe
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Cardiology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
19
|
Kawashima C, Matsuzawa Y, Akiyama E, Konishi M, Suzuki H, Hashiba K, Ebina T, Kosuge M, Hibi K, Tsukahara K, Iwahashi N, Maejima N, Sakamaki K, Umemura S, Kimura K, Tamura K. Prolonged Fever After ST-Segment Elevation Myocardial Infarction and Long-Term Cardiac Outcomes. J Am Heart Assoc 2017; 6:JAHA.116.005463. [PMID: 28735289 PMCID: PMC5586283 DOI: 10.1161/jaha.116.005463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background The biphasic inflammation after ST‐segment elevation myocardial infarction (STEMI) plays an important role in myocardial healing and progression of systemic atherosclerosis. The purpose of this study is to investigate the impact of fever during the first and second phases of post‐STEMI inflammation on long‐term cardiac outcomes. Methods and Results A total of 550 patients with STEMI were enrolled in this study. Axillary body temperature (BT) was measured and maximum BTs were determined for the first (within 3 days: max‐BT1–3d) and second (from 4 to 10 days after admission: max‐BT4–10d) phases, respectively. Patients were followed for cardiac events (cardiovascular death, acute coronary syndrome, and rehospitalization for heart failure) for a median 5.3 years. During the follow‐up period, 80 patients experienced cardiac events. A high max‐BT4–10d was strongly associated with long‐term cardiac events (hazard ratio, 95% CI) for a 1°C increase in the max‐BT4–10d: 2.834 (2.017–3.828), P<0.0001, whereas the max‐BT1–3d was not associated with cardiac events (1.136 [0.731–1.742], P=0.57). Even after adjustment for coronary risk factors, estimated glomerular filtration rate, infarct size, pericardial effusion, and medications on discharge, fever during the second phase (max‐BT4–10d ≥37.1°C) was significantly associated with future cardiac events (hazard ratio [95% CI] 2.900 [1.710–5.143], P<0.0001). Conclusions Fever during the second phase but not the first phase of post‐STEMI inflammation was a strong associated factor with worse long‐term cardiac outcomes in patients after STEMI, suggesting the need to consider the optimal timing for anti‐inflammatory strategies after STEMI.
Collapse
Affiliation(s)
- Chika Kawashima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Eiichi Akiyama
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaaki Konishi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Hiroyuki Suzuki
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Katsutaka Hashiba
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kengo Tsukahara
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Nobuhiko Maejima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kentaro Sakamaki
- Department of Biostatistics and Epidemiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Satoshi Umemura
- Division of Cardiology, Yokohama Rosai Hospital, Yokohama, Japan
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
20
|
Epicardial adipose tissue is associated with high-risk plaque feature progression in non-culprit lesions. Int J Cardiovasc Imaging 2017; 33:2029-2037. [PMID: 28550587 DOI: 10.1007/s10554-017-1158-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/26/2016] [Indexed: 10/19/2022]
Abstract
Several studies have suggested a link between epicardial adipose tissue (EAT) volume and high-risk plaques (HRPs); however, little is known about the association between EAT volume and HRP progression in non-culprit lesions (NCLs). Therefore, we evaluated whether EAT can independently predict HRP characteristic progression in NCLs by coronary computed tomography angiography (CCTA). In this single-centre trial, we analysed 131 consecutive patients (median age 61 years, male 72.52%) undergoing CCTA with percutaneous coronary intervention (PCI) for culprit lesions. All patients were scheduled to undergo follow-up CCTA 12 months after PCI. HRP features, including positive remodelling, low attenuation plaque, spotty calcification, and napkin-ring sign, along with EAT volume, were assessed by CCTA. The numbers of HRP features were compared between baseline and follow-up CCTA to detect HRP progression in NCLs, and patients were classified into two groups based on HRP progression. Logistic regression analysis was used to evaluate whether EAT volume was independently associated with HRP progression in NCLs. Overall, 23 of 131 patients who underwent two CCTAs exhibited HRP progression in NCLs (17.6%). Logistic regression analysis showed that a higher baseline EAT volume was associated with NCL HRP progression (odds ratio 1.019, 95% confidence interval 1.009-1.029, P < 0.001). The cut-off value for baseline EAT volume for NCL HRP progression was 107 ml based on receiver-operator characteristic curve analyses, and the area under the curve was 0.66. Baseline EAT volume was identified as an independent predictor of NCL HRP progression.
Collapse
|
21
|
Tan Y, Zhou J, Zhou Y, Yang X, Yang J, Chen Y. Characteristics Detected on Computed Tomography Angiography Predict Coronary Artery Plaque Progression in Non-Culprit Lesions. Korean J Radiol 2017; 18:487-497. [PMID: 28458601 PMCID: PMC5390618 DOI: 10.3348/kjr.2017.18.3.487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/03/2016] [Indexed: 11/15/2022] Open
Abstract
Objective This study sought to determine whether variables detected on coronary computed tomography angiography (CCTA) would predict plaque progression in non-culprit lesions (NCL). Materials and Methods In this single-center trial, we analyzed 103 consecutive patients who were undergoing CCTA and percutaneous coronary intervention (PCI) for culprit lesions. Follow-up CCTA was scheduled 12 months after the PCI, and all patients were followed for 3 years after their second CCTA examination. High-risk plaque features and epicardial adipose tissue (EAT) volume were assessed by CCTA. Each NCL stenosis grade was compared visually between two CCTA scans to detect plaque progression, and patients were stratified into two groups based on this. Logistic regression analysis was used to evaluate the factors that were independently associated with plaque progression in NCLs. Time-to-event curves were compared using the log-rank statistic. Results Overall, 34 of 103 patients exhibited NCL plaque progression (33%). Logistic regression analyses showed that the NCL progression was associated with a history of ST-elevated myocardial infarction (odds ratio [OR] = 5.855, 95% confidence interval [CI] = 1.391–24.635, p = 0.016), follow-up low-density lipoprotein cholesterol level (OR = 6.832, 95% CI = 2.103–22.200, p = 0.001), baseline low-attenuation plaque (OR = 7.311, 95% CI = 1.242–43.028, p = 0.028) and EAT (OR = 1.015, 95% CI = 1.000–1.029, p = 0.044). Following the second CCTA examination, major adverse cardiac events (MACEs) were observed in 12 patients, and NCL plaque progression was significantly associated with future MACEs (log rank p = 0.006). Conclusion Noninvasive assessment of NCLs by CCTA has potential prognostic value.
Collapse
Affiliation(s)
- Yahang Tan
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China.,The School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China.,Department of Cardiology, Tianjin Chest Hospital, Tianjin 300000, China
| | - Ying Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China.,Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiaobo Yang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China.,The School of Medicine, Nankai University, Tianjin 300071, China
| | - Junjie Yang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
22
|
Dutta P, Hoyer FF, Sun Y, Iwamoto Y, Tricot B, Weissleder R, Magnani JL, Swirski FK, Nahrendorf M. E-Selectin Inhibition Mitigates Splenic HSC Activation and Myelopoiesis in Hypercholesterolemic Mice With Myocardial Infarction. Arterioscler Thromb Vasc Biol 2016; 36:1802-8. [PMID: 27470513 DOI: 10.1161/atvbaha.116.307519] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/15/2016] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Atherosclerosis is a chronic disease characterized by lipid accumulation in the arterial wall. After myocardial infarction (MI), atherosclerotic plaques are infiltrated by inflammatory myeloid cells that aggravate the disease and increase the risk of secondary myocardial ischemia. Splenic myelopoiesis provides a steady flow of myeloid cells to inflamed atherosclerotic lesions after MI. Therefore, targeting myeloid cell production in the spleen could ameliorate increased atherosclerotic plaque inflammation after MI. APPROACH AND RESULTS Here we show that MI increases splenic myelopoiesis by driving hematopoietic stem and progenitor cells into the cell cycle. In an atherosclerotic mouse model, E-selectin inhibition decreased hematopoietic stem and progenitor cell proliferation in the spleen after MI. This led to reduced extramedullary myelopoiesis and decreased myeloid cell accumulation in atherosclerotic lesions. Finally, we observed stable atherosclerotic plaque features, including smaller plaque size, reduced necrotic core area, and thicker fibrous cap after E-selectin inhibition. CONCLUSIONS Inhibiting E-selectin attenuated inflammation in atherosclerotic plaques, likely by reducing leukocyte recruitment into plaques and by mitigating hematopoietic stem and progenitor cell activation in the spleen of mice with MI.
Collapse
Affiliation(s)
- Partha Dutta
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Friedrich Felix Hoyer
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Yuan Sun
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Yoshiko Iwamoto
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Benoit Tricot
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Ralph Weissleder
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - John L Magnani
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Filip K Swirski
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston (P.D., F.F.H., Y.S., Y.I., B.T., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and GlycoMimetics Inc, Rockville, MD (J.L.M.).
| |
Collapse
|
23
|
Han TW, Zhou SS, Li JT, Tian F, Mu Y, Jing J, Han YF, Chen YD. Homocysteine is associated with the progression of non-culprit coronary lesions in elderly acute coronary syndrome patients after percutaneous coronary intervention. J Geriatr Cardiol 2016; 13:299-305. [PMID: 27403138 PMCID: PMC4921541 DOI: 10.11909/j.issn.1671-5411.2016.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 02/26/2016] [Accepted: 02/27/2016] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The influence of homocysteine (Hcy) on the migration and proliferation of vascular smooth muscle cells has been well established. However, the impact of Hcy levels on the progression of non-culprit coronary lesions (NCCLs) is controversial. This study aims to evaluate whether the plasma level of Hcy is related to the progression of NCCLs after percutaneous coronary stent implantation in elderly patients with acute coronary syndrome (ACS). METHODS A total of 223 elderly patients (≥ 65 years old) with ACS undergoing stent implantation and follow-up coronary angiography were enrolled. Laboratory determination comprised of blood sample evaluation for Hcy was carried out before baseline coronary intervention. The patients were classified into two groups according to the blood Hcy tertiles (≥ 15 mmol/L or < 15 mmol/L). Patients were followed up for 12.2 months. NCCL progression was assessed by three-dimensional quantitative coronary angiography. RESULTS A significantly higher ratio of NCCL progression was observed in the group with baseline Hcy concentrations above 15 mmol/L compared to the group with concentrations below 15 mmol/L (41/127, 32.3% vs. 14/96, 14.6%, P = 0.002). Multivariate Cox regression analysis showed that Hcy and diabetes mellitus were independent risk factors for NCCL progression. The crude hazard ratio (HR) of NCCL progression for Hcy level was 1.056 (95% CI: 1.01-1.104, P = 0.015). The adjusted HR of NCCL progression for Hcy level was 1.024 (95% CI: 1.007-1.042, P = 0.007). The adjusted HR of NCCL progression for diabetes mellitus was 1.992 (95% CI: 1.15-3.44, P = 0.013). CONCLUSIONS Hcy is an independent risk factor for NCCL progression after 12 months of follow-up in elderly patients with ACS who has undergone percutaneous coronary stenting.
Collapse
Affiliation(s)
- Tian-Wen Han
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shan-Shan Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jian-Tao Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yang Mu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jing Jing
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yun-Feng Han
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yun-Dai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
24
|
Koskinas KC, Zaugg S, Yamaji K, García-García HM, Taniwaki M, Klingenberg R, Moschovitis A, Lüscher TF, van Tits LJ, Matter CM, Windecker S, Räber L. Changes of coronary plaque composition correlate with C-reactive protein levels in patients with ST-elevation myocardial infarction following high-intensity statin therapy. Atherosclerosis 2016; 247:154-60. [PMID: 26921743 DOI: 10.1016/j.atherosclerosis.2016.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/02/2016] [Accepted: 02/12/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Levels of inflammatory biomarkers associate with changes of coronary atheroma burden in statin-treated patients with stable coronary artery disease. This study sought to determine changes of plaque composition in vivo in relation to high-sensitivity C-reactive protein (hs-CRP) levels in patients with ST-elevation myocardial infarction (STEMI) receiving high-intensity statin therapy. METHODS The IBIS-4 study performed serial (baseline and 13-month), 2-vessel intravascular ultrasound (IVUS) and radiofrequency-IVUS of the non-infarct-related arteries in patients with STEMI treated with high-intensity statin therapy. The present analysis included 44 patients (80 arteries) with serial measurements of hs-CRP. RESULTS At follow-up, median low-density lipoprotein cholesterol (LDL-C) levels decreased from 126 to 77 mg/dl, HDL-C increased from 44 to 47 mg/dl, and hs-CRP decreased from 1.6 to 0.7 mg/L. Regression of percent atheroma volume (-0.99%, 95% CI -1.84 to -0.14, p = 0.024) was accompanied by reduction of percent fibro-fatty (p = 0.04) and fibrous tissue (p < 0.001), and increase in percent necrotic core (p = 0.006) and dense calcium (p < 0.001). Follow-up levels of hs-CRP, but not LDL-C, correlated with changes in percent necrotic core (p = 0.001) and inversely with percent fibrous tissue volume (p = 0.008). Similarly, baseline-to-follow-up change of hs-CRP correlated with the change in percent necrotic core volume (p = 0.02). CONCLUSIONS In STEMI patients receiving high-intensity statin therapy, stabilization of VH-IVUS-defined necrotic core was confined to patients with lowest on-treatment levels and greatest reduction of hs-CRP. Elevated CRP levels at follow-up may identify progression of high-risk coronary plaque composition despite intensive statin therapy and overall regression of atheroma volume.
Collapse
Affiliation(s)
| | - Serge Zaugg
- Clinical Trials Unit, Bern University, Bern, Switzerland
| | - Kyohei Yamaji
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | | | - Masanori Taniwaki
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | | | - Aris Moschovitis
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Thomas F Lüscher
- Cardiology Department, University Hospital Zurich, Zurich, Switzerland
| | | | | | - Stephan Windecker
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
25
|
Zhou Y, Tian F, Wang J, Yang JJ, Zhang T, Jing J, Chen YD. Efficacy study of olmesartan medoxomil on coronary atherosclerosis progression and epicardial adipose tissue volume reduction in patients with coronary atherosclerosis detected by coronary computed tomography angiography: study protocol for a randomized controlled trial. Trials 2016; 17:10. [PMID: 26739013 PMCID: PMC4702357 DOI: 10.1186/s13063-015-1097-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 12/01/2015] [Indexed: 02/01/2023] Open
Abstract
Background Epicardial adipose tissue (EAT) is a newly discovered independent risk factor for coronary atherosclerosis. There is a scarcity of information on the reduction of EAT volume to reduce atherosclerosis risk. Coronary computed tomography angiography (CCTA) has emerged as a noninvasive imaging method for the analysis of coronary atherosclerosis and EAT volume. The purpose of this trial is to determine whether olmesartan medoxomil is effective at both treatment of coronary atherosclerosis progression and EAT volume reduction in patients with coronary atherosclerosis detected by CCTA. Methods/design This study is a prospective, single-center, open-label, randomized controlled clinical trial aimed at exploring the efficacy of olmesartan medoxomil on coronary atherosclerosis and EAT. A total of 194 patients with coronary stenosis greater than 30 % and less than 70 % detected by CCTA will be randomly divided into olmesartan medoxomil or conventional antihypertensive medication groups (1:1 ratio). The primary outcome measures include coronary atherosclerosis progression and EAT volume reduction, as detected by CCTA at 12 months. The secondary outcome measures include the levels of blood lipids, glucose, high-sensitivity C-reactive protein, IL-6, monocyte chemotactic protein 1, TNF-α, matrix metalloproteinase 9, NO, endothelin 1, adiponectin, and leptin at baseline and after 6 and 12 months. Discussion Treatments aimed at reducing EAT volume can eventually achieve an antiatherosclerotic effect. This is the first trial designed to explore the effect of olmesartan medoxomil on both coronary atherosclerosis progression and EAT volume reduction in patients with coronary atherosclerosis detected by CCTA. Trial registration ClinicalTrials.gov: NCT02360956.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jing Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jun-Jie Yang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Tao Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jing Jing
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yun-Dai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
26
|
Kugathasan P, Aagaard J, Jensen S. Coronary artery disease as a cause of morbidity and mortality in patients suffering from schizophrenia: protocol for a prospective cohort study with long-term follow-up. ACTA ACUST UNITED AC 2016. [DOI: 10.4103/2468-5658.196979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
27
|
Nahrendorf M, Swirski FK. Innate immune cells in ischaemic heart disease: does myocardial infarction beget myocardial infarction? Eur Heart J 2015; 37:868-72. [PMID: 26351395 DOI: 10.1093/eurheartj/ehv453] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/17/2015] [Indexed: 12/24/2022] Open
Abstract
Knowledge of macrophages in steady-state and diseased tissue is rapidly expanding, propelled by improved diagnostic capacity to detect and monitor cells in their native environments. In this review, we discuss implications for ischaemic heart disease and examine innate immune cell pathways that increase systemic leucocyte supply after myocardial infarction (MI). Acute MI alters the macrophage phenotype and supply chain from tissue resident to blood monocytes sourced from haematopoietic organs. That blood leucocytosis closely associates with cardiovascular mortality provides a strong motivation to understand why and how organ ischaemia alters cellular immunity.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
28
|
Joshi NV, Toor I, Shah ASV, Carruthers K, Vesey AT, Alam SR, Sills A, Hoo TY, Melville AJ, Langlands SP, Jenkins WSA, Uren NG, Mills NL, Fletcher AM, van Beek EJR, Rudd JHF, Fox KAA, Dweck MR, Newby DE. Systemic Atherosclerotic Inflammation Following Acute Myocardial Infarction: Myocardial Infarction Begets Myocardial Infarction. J Am Heart Assoc 2015; 4:e001956. [PMID: 26316523 PMCID: PMC4599491 DOI: 10.1161/jaha.115.001956] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preclinical data suggest that an acute inflammatory response following myocardial infarction (MI) accelerates systemic atherosclerosis. Using combined positron emission and computed tomography, we investigated whether this phenomenon occurs in humans. METHODS AND RESULTS Overall, 40 patients with MI and 40 with stable angina underwent thoracic 18F-fluorodeoxyglucose combined positron emission and computed tomography scan. Radiotracer uptake was measured in aortic atheroma and nonvascular tissue (paraspinal muscle). In 1003 patients enrolled in the Global Registry of Acute Coronary Events, we assessed whether infarct size predicted early (≤30 days) and late (>30 days) recurrent coronary events. Compared with patients with stable angina, patients with MI had higher aortic 18F-fluorodeoxyglucose uptake (tissue-to-background ratio 2.15±0.30 versus 1.84±0.18, P<0.0001) and plasma C-reactive protein concentrations (6.50 [2.00 to 12.75] versus 2.00 [0.50 to 4.00] mg/dL, P=0.0005) despite having similar aortic (P=0.12) and less coronary (P=0.006) atherosclerotic burden and similar paraspinal muscular 18F-fluorodeoxyglucose uptake (P=0.52). Patients with ST-segment elevation MI had larger infarcts (peak plasma troponin 32 300 [10 200 to >50 000] versus 3800 [1000 to 9200] ng/L, P<0.0001) and greater aortic 18F-fluorodeoxyglucose uptake (2.24±0.32 versus 2.02±0.21, P=0.03) than those with non-ST-segment elevation MI. Peak plasma troponin concentrations correlated with aortic 18F-fluorodeoxyglucose uptake (r=0.43, P=0.01) and, on multivariate analysis, independently predicted early (tertile 3 versus tertile 1: relative risk 4.40 [95% CI 1.90 to 10.19], P=0.001), but not late, recurrent MI. CONCLUSIONS The presence and extent of MI is associated with increased aortic atherosclerotic inflammation and early recurrent MI. This finding supports the hypothesis that acute MI exacerbates systemic atherosclerotic inflammation and remote plaque destabilization: MI begets MI. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01749254.
Collapse
Affiliation(s)
- Nikhil V Joshi
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Iqbal Toor
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Anoop S V Shah
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Kathryn Carruthers
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Alex T Vesey
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Shirjel R Alam
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Andrew Sills
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Teng Y Hoo
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Adam J Melville
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Sarah P Langlands
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - William S A Jenkins
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Neal G Uren
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Alison M Fletcher
- Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.)
| | - Edwin J R van Beek
- Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.)
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (J.F.R.)
| | - Keith A A Fox
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., A.S., T.Y.H., A.J.M., S.P.L., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.) Clinical Research Imaging Centre, University of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., A.M.F., E.R.B., K.A.F., M.R.D., D.E.N.) Edinburgh Heart Centre, Royal Infirmary of Edinburgh, United Kingdom (N.V.J., I.T., A.V.S., K.C., A.T.V., S.R.A., W.A.J., N.G.U., N.L.M., K.A.F., M.R.D., D.E.N.)
| |
Collapse
|
29
|
Park CS, Chung WB, Choi YS, Kim PJ, Lee JM, Baek KH, Kim HY, Yoo KD, Song KH, Chung WS, Seung KB, Lee MY, Kwon HS. Acute Myocardial Infarction Is a Risk Factor for New Onset Diabetes in Patients with Coronary Artery Disease. PLoS One 2015; 10:e0136354. [PMID: 26295946 PMCID: PMC4546589 DOI: 10.1371/journal.pone.0136354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/01/2015] [Indexed: 01/04/2023] Open
Abstract
Objective To test the hypothesis that acute myocardial infarction (AMI) might accelerate development of new onset diabetes in patients with coronary artery disease independent of known risk factors. Methods We conducted a retrospective cohort study within COACT (CathOlic medical center percutAneous Coronary inTervention) registry. From a total of 9,127 subjects, 2,036 subjects were diabetes naïve and followed up for at least one year with both index and follow-up laboratory data about diabetes. Cox proportional hazard model was used to derive hazard ratios (HRs) and 95% confidence interval (CI) for new onset diabetes associated with AMI in univariate and multivariate analysis after adjusting several covariates. Results The overall hazard for diabetes was higher in AMI compared to non-AMI patients (p by log rank <0.01) with HR of 1.78 and 95% CI of 1.37–2.32 in univariate analysis. This association remained significant after adjusting covariates (HR, 1.54; 95% CI, 1.14–2.07; p<0.01). AMI was an independent predictor for higher quartile of WBC count in multivariate ordinal logistic regression analysis (OR, 6.75; 95% CI, 5.53–8.22, p<0.01). In subgroup analysis, the diabetogenic effect of AMI was more prominent in the subgroup without MetS compared to MetS patients (p for interaction<0.05). Compared to the reference group of non-AMI+nonMetS, the group of AMI+non-MetS (HR, 2.44; 95% CI, 1.58–3.76), non-AMI+MetS (HR, 3.42; 95% CI, 2.34–4.98) and AMI+MetS (HR, 4.12; 95% CI, 2.67–6.36) showed higher HR after adjusting covariates. However, the hazard was not different between the non-AMI+MetS and AMI+non-MetS groups. Conclusions AMI patients have a greater risk of new-onset diabetes when compared to non AMI patients, especially those with mild metabolic abnormalities.
Collapse
Affiliation(s)
- Chul Soo Park
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woo Baek Chung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yun Seok Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Pum Joon Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Seoul St. Mary’s Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Min Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Ki-Hyun Baek
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee Yeol Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Bucheon St. Mary’s Hospital, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Ki Dong Yoo
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, St. Vincent Hospital, The Catholic University of Korea, Suwon, Republic of Korea
| | - Ki-Ho Song
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wook Sung Chung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Seoul St. Mary’s Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Ki Bae Seung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Seoul St. Mary’s Hospital, Catholic University of Korea, Seoul, Republic of Korea
| | - Man Young Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Center and Cardiology Division, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Sang Kwon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
30
|
Abstract
Monocytes are part of the vertebrate innate immune system. Blood monocytes are produced by bone marrow and splenic progenitors that derive from hematopoietic stem cells (HSCs). In cardiovascular disease, such as atherosclerosis and myocardial infarction, HSCs proliferate at higher levels that in turn increase production of hematopoietic cells, including monocytes. Once produced in hematopoietic niches, monocytes intravasate blood vessels, circulate, and migrate to sites of inflammation. Monocyte recruitment to atherosclerotic plaque and the ischemic heart depends on various chemokines, such as CCL2, CX3 CL1, and CCL5. Once in tissue, monocytes can differentiate into macrophages and dendritic cells. Macrophages are end effector cells that regulate the steady state and tissue healing, but they can also promote disease. At sites of inflammation, monocytes and macrophages produce inflammatory cytokines, which can exacerbate disease progression. Macrophages can also phagocytose tissue debris and produce pro-healing cytokines. Additionally, macrophages are antigen-presenting cells and can prime T cells. The tissue environment, including cytokines and types of inflammation, instructs macrophage specialization. Understanding monocytosis and its consequences in disease will reveal new therapeutic opportunities without compromising steady state functions.
Collapse
Affiliation(s)
- Partha Dutta
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
31
|
Dutta P, Nahrendorf M. Monocytes in myocardial infarction. Arterioscler Thromb Vasc Biol 2015; 35:1066-70. [PMID: 25792449 PMCID: PMC4409536 DOI: 10.1161/atvbaha.114.304652] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/27/2015] [Indexed: 01/06/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death in developed countries. Though timely revascularization of the ischemic myocardium and current standard therapy reduce acute mortality after MI, long-term morbidity and mortality remain high. During the first 1 to 2 weeks after MI, tissues in the infarcted myocardium undergo rapid turnover, including digestion of extracellular matrix and fibrosis. Post-MI repair is crucial to survival. Monocytes recruited to the infarcted myocardium remove debris and facilitate the repair process. However, exaggerated inflammation may also impede healing, as demonstrated by the association between elevated white blood cell count and in-hospital mortality after MI. Monocytes produced in the bone marrow and spleen enter the blood after MI and are recruited to the injured myocardium in 2 phases. The first phase is dominated by Ly-6c(high) monocytes and the second phase by Ly-6c(low) monocytes. Yet the number of Ly6C(low) monocytes recruited to the infarct is much lower, and Ly6C(high) monocytes can differentiate to Ly6C(low) macrophages in later healing stages. Understanding the signals regulating monocytosis after MI will help design new therapies to facilitate cardiac healing and limit heart failure.
Collapse
Affiliation(s)
- Partha Dutta
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
32
|
Tang J, Lobatto ME, Hassing L, van der Staay S, van Rijs SM, Calcagno C, Braza MS, Baxter S, Fay F, Sanchez-Gaytan BL, Duivenvoorden R, Sager HB, Astudillo YM, Leong W, Ramachandran S, Storm G, Pérez-Medina C, Reiner T, Cormode DP, Strijkers GJ, Stroes ESG, Swirski FK, Nahrendorf M, Fisher EA, Fayad ZA, Mulder WJM. Inhibiting macrophage proliferation suppresses atherosclerotic plaque inflammation. SCIENCE ADVANCES 2015; 1:e1400223. [PMID: 26295063 PMCID: PMC4539616 DOI: 10.1126/sciadv.1400223] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/05/2015] [Indexed: 05/29/2023]
Abstract
Inflammation drives atherosclerotic plaque progression and rupture, and is a compelling therapeutic target. Consequently, attenuating inflammation by reducing local macrophage accumulation is an appealing approach. This can potentially be accomplished by either blocking blood monocyte recruitment to the plaque or increasing macrophage apoptosis and emigration. Because macrophage proliferation was recently shown to dominate macrophage accumulation in advanced plaques, locally inhibiting macrophage proliferation may reduce plaque inflammation and produce long-term therapeutic benefits. To test this hypothesis, we used nanoparticle-based delivery of simvastatin to inhibit plaque macrophage proliferation in apolipoprotein E deficient mice (Apoe-/- ) with advanced atherosclerotic plaques. This resulted in rapid reduction of plaque inflammation and favorable phenotype remodeling. We then combined this short-term nanoparticle intervention with an eight-week oral statin treatment, and this regimen rapidly reduced and continuously suppressed plaque inflammation. Our results demonstrate that pharmacologically inhibiting local macrophage proliferation can effectively treat inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Jun Tang
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark E. Lobatto
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Laurien Hassing
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Susanne van der Staay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Sarian M. van Rijs
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mounia S. Braza
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Samantha Baxter
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Francois Fay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brenda L. Sanchez-Gaytan
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raphaël Duivenvoorden
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Hendrik B. Sager
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yaritzy M. Astudillo
- Department of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Wei Leong
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarayu Ramachandran
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, Netherlands
- Department of Controlled Drug Delivery, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, 7500 AE Enschede, Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gustav J. Strijkers
- Department of Biomedical Engineering and Physics, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| | - Filip K. Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Edward A. Fisher
- Department of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Zahi A. Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Willem J. M. Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, Netherlands
| |
Collapse
|
33
|
Non-culprit coronary lesions in young patients have higher rates of atherosclerotic progression. Int J Cardiovasc Imaging 2015; 31:889-97. [DOI: 10.1007/s10554-015-0635-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
|
34
|
Cardiovascular imaging 2014 in the International Journal of Cardiovascular Imaging. Int J Cardiovasc Imaging 2015; 31:447-61. [DOI: 10.1007/s10554-015-0627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
35
|
Abstract
Congestive heart failure (CHF) remains a significant cause of death and disability in industrialized countries. Projections show that the prevalence of CHF will increase 46% from 2012 to 2030, resulting in over eight million adults with CHF in the United States. While substantial advances have been achieved in the treatment of CHF over the past two decades, CHF rivals cancer as a cause of mortality. Strategies focused on prevention of CHF should be emphasized to meaningfully impact the projected increase in CHF. Irrespective of the type of CHF, either systolic or diastolic, coronary artery disease has supplanted hypertension as the most prevalent cause for congestive heart failure, with a high rate of mortality and future hospitalizations. Since coronary artery disease plays a central role in the development of CHF, approaches to treat coronary artery disease and identification of patients at risk for recurrent myocardial infarction (RMI) are approaches to prevent development of CHF. Subjects who sustain recurrent MI represent a particularly high-risk group for development of CHF. Despite the evolution of therapy for MI from thrombolytic therapy to primary percutaneous coronary intervention (PCI), RMI occurs in ~ 10% of patients in the first year after first MI, and 3 years after their first MI. In this review I explore emerging approaches to prevent RMI including the rationale for recent trials of complete revascularization at the time of MI, newly emerging biomarkers that have additive predictive value for identifying patients with high risk of CHF and death when using existing biomarkers. Finally, the paradigm of hematopoietic stem cell mobilization in MI leading to monocyte expansion and acceleration of atherosclerosis is discussed as an emerging approach to identify patients at high risk of RMI, CHF, and death after MI.
Collapse
|