1
|
Şaylık F, Çınar T, Selçuk M, Akbulut T, Hayıroğlu Mİ, Tanboğa İH. Evaluation of Naples Score for Long-Term Mortality in Patients With ST-Segment Elevation Myocardial Infarction Undergoing Primary Percutaneous Coronary Intervention. Angiology 2024; 75:725-733. [PMID: 37058422 DOI: 10.1177/00033197231170982] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The Naples score (NS), which is a composite of cardiovascular adverse event predictors including neutrophil-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, albumin, and total cholesterol, has emerged as a prognostic risk score in cancer patients. We aimed to investigate the predictive value of NS for long-term mortality in ST-segment elevation myocardial infarction patients (STEMI). A total of 1889 STEMI patients were enrolled in this study. The median duration of the study was 43 months (IQR: 32-78). Patients were divided into 2 groups according to NS as group 1 and group 2. We created 3 models as a baseline model, model 1 (baseline + NS in continuous), and model 2 (baseline + NS as categorical). Group 2 patients had higher long-term mortality rates than group 1 patients. The NS was independently associated with long-term mortality and adding NS to a baseline model improved the model performance for prediction and discrimination of long-term mortality. Decision curve analysis demonstrated that model 1 had a better net benefit probability for detecting mortality compared with the baseline model. NS had the highest contributive significant effect in the prediction model. An easily accessible and calculable NS might be used for risk stratification of long-term mortality in STEMI patients undergoing primary percutaneous coronary intervention.
Collapse
Affiliation(s)
- Faysal Şaylık
- Department of Cardiology, Van Education and Research Hospital, Van, Turkey
| | - Tufan Çınar
- Department of Cardiology, Sultan Abdulhamid Han Education and Research Hospital, Istanbul, Turkey
| | - Murat Selçuk
- Department of Cardiology, Sultan Abdulhamid Han Education and Research Hospital, Istanbul, Turkey
| | - Tayyar Akbulut
- Department of Cardiology, Van Education and Research Hospital, Van, Turkey
| | - Mert İlker Hayıroğlu
- Department of Cardiology, Dr. Siyami Ersek Education and Research Hospital, Istanbul, Turkey
| | | |
Collapse
|
2
|
Berna-Rico E, Abbad-Jaime de Aragon C, Ballester-Martinez A, Perez-Bootello J, Solis J, Fernandez-Friera L, Llamas-Velasco M, Castellanos-Gonzalez M, Barderas MG, Azcarraga-Llobet C, Garcia-Mouronte E, de Nicolas-Ruanes B, Naharro-Rodriguez J, Jaen-Olasolo P, Gelfand JM, Mehta NN, Gonzalez-Cantero A. Monocyte-to-High-Density Lipoprotein Ratio Is Associated with Systemic Inflammation, Insulin Resistance, and Coronary Subclinical Atherosclerosis in Psoriasis: Results from 2 Observational Cohorts. J Invest Dermatol 2024; 144:2002-2012.e2. [PMID: 38460808 DOI: 10.1016/j.jid.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/11/2024]
Abstract
Systemic inflammation or insulin resistance drive atherosclerosis. However, they are difficult to capture for assessing cardiovascular risk in clinical settings. The monocyte-to-high-density lipoprotein ratio (MHR) is an accessible biomarker that integrates inflammatory and metabolic information and has been associated with poorer cardiovascular outcomes. Our aim was to evaluate the association of MHR with the presence of subclinical atherosclerosis in patients with psoriasis. The study involved a European and an American cohort including 405 patients with the disease. Subclinical atherosclerosis was assessed by coronary computed tomography angiography. First, MHR correlated with insulin resistance through homeostatic model assessment for insulin resistance, with high-sensitivity CRP and with 18F-fluorodeoxyglucose uptake in spleen, liver, and bone marrow by positron emission tomography/computed tomography. MHR was associated with both the presence of coronary plaques >50% of the artery lumen and noncalcified coronary burden, beyond traditional cardiovascular risk factors (P < .05). In a noncalcified coronary burden prediction model accounting for cardiovascular risk factors, statins, and biologic treatment, MHR added value (area under the curve base model = 0.72 vs area under the curve base model plus MHR = 0.76, P = .04) within the American cohort. These results suggests that MHR may detect patients with psoriasis who have subclinical burden of cardiovascular disease and warrant more aggressive measures to reduce lifetime adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Emilio Berna-Rico
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Carlota Abbad-Jaime de Aragon
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Asuncion Ballester-Martinez
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Perez-Bootello
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jorge Solis
- Department of Cardiology, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Cardiology, Atria Clinic, Madrid, Spain; Centro Integral de Enfermedades Cardiovasculares (CIEC), Hospital Universitario HM Montepríncipe, HM Hospitales, Madrid, Spain; Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Leticia Fernandez-Friera
- Department of Cardiology, Atria Clinic, Madrid, Spain; Centro Integral de Enfermedades Cardiovasculares (CIEC), Hospital Universitario HM Montepríncipe, HM Hospitales, Madrid, Spain; Facultad HM Hospitales de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Mar Llamas-Velasco
- Department of Dermatology, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha (SESCAM), Toledo, Spain
| | - Carlos Azcarraga-Llobet
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Emilio Garcia-Mouronte
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Belen de Nicolas-Ruanes
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jorge Naharro-Rodriguez
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pedro Jaen-Olasolo
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Joel M Gelfand
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nehal N Mehta
- Department of Cardiology, George Washington Medical Center, Washington, District of Columbia, USA
| | - Alvaro Gonzalez-Cantero
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain.
| |
Collapse
|
3
|
Chen M, Wu Q, Shao N, Lai X, Lin H, Chen M, Wu Y, Chen J, Lin Q, Huang J, Chen X, Yan W, Chen S, Li H, Wu D, Yang M, Deng C. The significance of CD16+ monocytes in the occurrence and development of chronic thromboembolic pulmonary hypertension: insights from single-cell RNA sequencing. Front Immunol 2024; 15:1446710. [PMID: 39192976 PMCID: PMC11347785 DOI: 10.3389/fimmu.2024.1446710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Background Chronic thromboembolic pulmonary hypertension (CTEPH) is a serious pulmonary vascular disease characterized by residual thrombi in the pulmonary arteries and distal pulmonary microvascular remodeling. The pathogenesis of CTEPH remains unclear, but many factors such as inflammation, immunity, coagulation and angiogenesis may be involved. Monocytes are important immune cells that can differentiate into macrophages and dendritic cells and play an important role in thrombus formation. However, the distribution, gene expression profile and differentiation trajectory of monocyte subsets in CTEPH patients have not been systematically studied. This study aims to reveal the characteristics and functions of monocytes in CTEPH patients using single-cell sequencing technology, and to provide new insights for the diagnosis and treatment of CTEPH. Methods Single-cell RNA sequencing (scRNA-seq) were performed to analyze the transcriptomic features of peripheral blood mononuclear cells (PBMCs) from healthy controls, CTEPH patients and the tissues from CTEPH patients after the pulmonary endarterectomy (PEA). We established a CTEPH rat model with chronic pulmonary embolism caused by repeated injection of autologous thrombi through a central venous catheter, and used flow cytometry to detect the proportion changes of monocyte subsets in CTEPH patients and CTEPH rat model. We also observed the infiltration degree of macrophage subsets in thrombus tissue and their differentiation relationship with peripheral blood monocyte subsets by immunofluorescence staining. Results The results showed that the monocyte subsets in peripheral blood of CTEPH patients changed significantly, especially the proportion of CD16+ monocyte subset increased. This monocyte subset had unique functional features at the transcriptomic level, involving processes such as cell adhesion, T cell activation, coagulation response and platelet activation, which may play an important role in pulmonary artery thrombus formation and pulmonary artery intimal remodeling. In addition, we also found that the macrophage subsets in pulmonary endarterectomy tissue of CTEPH patients showed pro-inflammatory and lipid metabolism reprogramming features, which may be related to the persistence and insolubility of pulmonary artery thrombi and the development of pulmonary hypertension. Finally, we also observed that CD16+ monocyte subset in peripheral blood of CTEPH patients may be recruited to pulmonary artery intimal tissue and differentiate into macrophage subset with high expression of IL-1β, participating in disease progression. Conclusion CD16+ monocytes subset had significant gene expression changes in CTEPH patients, related to platelet activation, coagulation response and inflammatory response. And we also found that these cells could migrate to the thrombus and differentiate into macrophages with high expression of IL-1β involved in CTEPH disease progression. We believe that CD16+ monocytes are important participants in CTEPH and potential therapeutic targets.
Collapse
Affiliation(s)
- Maohe Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qiuxia Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Nan Shao
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xingyue Lai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Huo Lin
- Department of Pulmonary and Critical Care Medicine, Shishi County Hospital, Shishi, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Yijing Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiafan Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qinghuang Lin
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiahui Huang
- Department of Respiratory and Critical Care Medicine, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Xiaoyun Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Wei Yan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Shi Chen
- Department of Respiratory and Critical Care, Wuhan No. 6 Hospital, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Hongli Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dawen Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Minxia Yang
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chaosheng Deng
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
4
|
Wang H, Rouhi N, Slotabec LA, Seale BC, Wen C, Filho F, Adenawoola MI, Li J. Myeloid Cells in Myocardial Ischemic Injury: The Role of the Macrophage Migration Inhibitory Factor. Life (Basel) 2024; 14:981. [PMID: 39202723 PMCID: PMC11355293 DOI: 10.3390/life14080981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Ischemic heart disease, manifesting as myocardial infarction (MI), remains the leading cause of death in the western world. Both ischemia and reperfusion (I/R) cause myocardial injury and result in cardiac inflammatory responses. This sterile inflammation in the myocardium consists of multiple phases, involving cell death, tissue remodeling, healing, and scar formation, modulated by various cytokines, including the macrophage migration inhibitory factor (MIF). Meanwhile, different immune cells participate in these phases, with myeloid cells acting as first responders. They migrate to the injured myocardium and regulate the initial phase of inflammation. The MIF modulates the acute inflammatory response by affecting the metabolic profile and activity of myeloid cells. This review summarizes the role of the MIF in regulating myeloid cell subsets in MI and I/R injury and discusses emerging evidence of metabolism-directed cellular inflammatory responses. Based on the multifaceted role of the MIF affecting myeloid cells in MI or I/R, the MIF can be a therapeutic target to achieve metabolic balance under pathology and alleviate inflammation in the heart.
Collapse
Affiliation(s)
- Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Lily A. Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, USA
| | - Blaise C. Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Michael I. Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
5
|
Wang X, Cheng H, Feng M, Jiang B, Ren C, Chen Q, Zhi X, Li Y. Causality of genetically proxied immunophenotypes on cardiovascular diseases: a Mendelian randomization study. Front Immunol 2024; 15:1344773. [PMID: 38887301 PMCID: PMC11181691 DOI: 10.3389/fimmu.2024.1344773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Background Cardiovascular diseases (CVDs) stand as the foremost global cause of mortality, prompting a growing interest in using the potential of immune cells for heart injury treatment. This study aims to assess the causal association between immune cells and CVDs. Methods A total of 731 immune cells were derived from a previously published genome-wide association study (GWAS), which included approximately 22 million genetic variants among 3,757 individuals of Sardinian ancestry. Genetic associations with atrial fibrillation (AF), heart failure, coronary artery disease, myocardial infarction and stroke were extracted from large-scale GWAS. A two-sample Mendelian randomization (MR) analysis was used to assess the causal association between immune cells and CVDs. Replication MR analysis based on FinnGen dataset and meta-analysis are sequentially conducted to validate causal relationships. Results Collectively, genetically predicted 4 immune cell traits were associated with AF and 5 immune cell traits were associated with stroke. Increased levels of IgD- CD38dim absolute count were associated with a higher susceptibility to AF, while increased expression of CD14+ CD16+ monocytes, CD62L on CD62L+ myeloid dendritic cells, and CD16 on CD14- CD16+ monocytes were linked to a decreased susceptibility to AF. Additionally, an elevated susceptibility to stroke was linked to an increase in the percentage of CD39+ resting Tregs and heightened CD27 expression on IgD- CD38+ cells. Conversely, a decreased susceptibility to stroke was associated with increased CD40 expression on monocytes, particularly on CD14+ CD16+ and CD14+ CD16- monocytes, with the latter two showing the most compelling evidence. Conclusion This study identified several immune cell traits that have a causal relationship with CVDs, thus confirming that immune cells play an important role in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Xuehan Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huixin Cheng
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Meng Feng
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bing Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yingdong Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Wang L, Gao J, Liu B, Fu Y, Yao Z, Guo S, Song Z, Zhang Z, He J, Wang C, Ma W, Wu F. The association between lymphocyte-to-monocyte ratio and all-cause mortality in obese hypertensive patients with diabetes and without diabetes: results from the cohort study of NHANES 2001-2018. Front Endocrinol (Lausanne) 2024; 15:1387272. [PMID: 38686205 PMCID: PMC11056572 DOI: 10.3389/fendo.2024.1387272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Objective Obesity, hypertension and diabetes are high prevalent that are often associated with poor outcomes. They have become major global health concern. Little research has been done on the impact of lymphocyte-to-monocyte ratio (LMR) on outcomes in these patients. Thus, we aimed to explore the association between LMR and all-cause mortality in obese hypertensive patients with diabetes and without diabetes. Methods The researchers analyzed data from the National Health and Nutrition Examination Survey (2001-2018), which included 4,706 participants. Kaplan-Meier analysis was employed to compare survival rate between different groups. Multivariate Cox proportional hazards regression models with trend tests and restricted cubic splines (RCS) analysis and were used to investigate the relationship between the LMR and all-cause mortality. Subgroup analysis was performed to assess whether there was an interaction between the variables. Results The study included a total of 4706 participants with obese hypertension (48.78% male), of whom 960 cases (20.40%) died during follow-up (median follow-up of 90 months). Kaplan-Meier curves suggested a remarkable decrease in all-cause mortality with increasing LMR value in patients with diabetes and non-diabetes (P for log-rank test < 0.001). Moreover, multivariable Cox models demonstrated that the risk of mortality was considerably higher in the lowest quartile of the LMR and no linear trend was observed (P > 0.05). Furthermore, the RCS analysis indicated a non-linear decline in the risk of death as LMR values increased (P for nonlinearity < 0.001). Conclusions Increased LMR is independently related with reduced all-cause mortality in patients with obese hypertension, regardless of whether they have combined diabetes.
Collapse
Affiliation(s)
- Lixia Wang
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Jie Gao
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Bing Liu
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Youliang Fu
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Zhihui Yao
- Department of Cardiology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shanshan Guo
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Ziwei Song
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Zhaoyuan Zhang
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Jiaojiao He
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Congxia Wang
- Department of Cardiology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weidong Ma
- Department of Cardiology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Feng Wu
- Department of Cardiology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
7
|
Yan J, Tie G, Tutto A, Messina LM. Hypercholesterolemia impairs collateral artery enlargement by ten-eleven translocation 1-dependent hematopoietic stem cell autonomous mechanism in a murine model of limb ischemia. JVS Vasc Sci 2024; 5:100203. [PMID: 38774713 PMCID: PMC11106542 DOI: 10.1016/j.jvssci.2024.100203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/22/2024] [Indexed: 05/24/2024] Open
Abstract
Objective The extent of collateral artery enlargement determines the risk of limb loss due to peripheral arterial disease. Hypercholesterolemia impairs collateral artery enlargement, but the underlying mechanism remains poorly characterized. This study tests the hypothesis that hypercholesterolemia impairs collateral artery enlargement through a ten-eleven translocation 1 (Tet1)-dependent hematopoietic stem cell (HSC)-autonomous mechanism that increases their differentiation into proinflammatory Ly6Chi monocytes and restricts their conversion into proangiogenic Ly6Clow monocytes. Methods To test our hypothesis, we induced limb ischemia and generated chimeric mouse models by transplanting HSCs from either wild-type (WT) mice or hypercholesterolemic mice into lethally irradiated WT recipient mice. Results We found that the lethally irradiated WT recipient mice reconstituted with HSCs from hypercholesterolemic mice displayed lower blood flow recovery and collateral artery enlargement that was nearly identical to that observed in hypercholesterolemic mice, despite the absence of hypercholesterolemia and consistent with an HSC-autonomous mechanism. We showed that hypercholesterolemia impairs collateral artery enlargement by a Tet1-dependent mechanism that increases HSC differentiation toward proinflammatory Ly6Chi monocytes and restricts the conversion of Ly6Chi monocytes into proangiogenic Ly6Clow monocytes. Moreover, Tet1 epigenetically reprograms monocyte gene expression within the HSCs. Restoration of Tet1 expression in HSCs of hypercholesterolemic mice restores WT collateral artery enlargement and blood flow recovery after induction of hindlimb ischemia. Conclusions These results show that hypercholesterolemia impairs collateral artery enlargement by a novel Tet1-dependent HSC-autonomous mechanism that epigenetically reprograms monocyte gene expression within the HSCs.
Collapse
Affiliation(s)
- Jinglian Yan
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Guodong Tie
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Amanda Tutto
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| | - Louis M. Messina
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
8
|
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. LAB ON A CHIP 2024; 24:1494-1528. [PMID: 38318723 DOI: 10.1039/d3lc00829k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, casting a substantial economic footprint and burdening the global healthcare system. Historically, pre-clinical CVD modeling and therapeutic screening have been performed using animal models. Unfortunately, animal models oftentimes fail to adequately mimic human physiology, leading to a poor translation of therapeutics from pre-clinical trials to consumers. Even those that make it to market can be removed due to unforeseen side effects. As such, there exists a clinical, technological, and economical need for systems that faithfully capture human (patho)physiology for modeling CVD, assessing cardiotoxicity, and evaluating drug efficacy. Heart-on-a-chip (HoC) systems are a part of the broader organ-on-a-chip paradigm that leverages microfluidics, tissue engineering, microfabrication, electronics, and gene editing to create human-relevant models for studying disease, drug-induced side effects, and therapeutic efficacy. These compact systems can be capable of real-time measurements and on-demand characterization of tissue behavior and could revolutionize the drug development process. In this review, we highlight the key components that comprise a HoC system followed by a review of contemporary reports of their use in disease modeling, drug toxicity and efficacy assessment, and as part of multi-organ-on-a-chip platforms. We also discuss future perspectives and challenges facing the field, including a discussion on the role that standardization is expected to play in accelerating the widespread adoption of these platforms.
Collapse
Affiliation(s)
- Derrick Butler
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | - Darwin R Reyes
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
9
|
Guo J, Huang Y, Pang L, Zhou Y, Yuan J, Zhou B, Fu M. Association of systemic inflammatory response index with ST segment elevation myocardial infarction and degree of coronary stenosis: a cross-sectional study. BMC Cardiovasc Disord 2024; 24:98. [PMID: 38336634 PMCID: PMC10858502 DOI: 10.1186/s12872-024-03751-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Systemic Inflammatory Response Index (SIRI), a composite inflammatory marker encompassing neutrophils, monocytes, and lymphocytes, has been recognized as a reliable marker of systemic inflammation. This article undertakes an analysis of clinical data from ST-segment Elevation Myocardial Infarction (STEMI) patients, aiming to comprehensively assess the relationship between SIRI, STEMI, and the degree of coronary stenosis. METHODS The study involved 1809 patients diagnosed with STEMI between the years 2020 and 2023. Univariate and multivariate logistic regression analyses were conducted to evaluate the risk factors for STEMI. Receiver operating characteristic (ROC) curves were generated to determine the predictive power of SIRI and neutrophil-to-lymphocyte ratio (NLR). Spearman correlation analysis was performed to assess the correlation between SIRI, NLR, and the Gensini score (GS). RESULTS Multivariate logistic regression analysis showed that the SIRI was the independent risk factor for STEMI (adjusted odds ratio (OR) in the highest quartile = 24.96, 95% confidence interval (CI) = 15.32-40.66, P < 0.001). In addition, there is a high correlation between SIRI and GS (β:28.54, 95% CI: 24.63-32.46, P < 0.001). The ROC curve analysis was performed to evaluate the predictive ability of SIRI and NLR for STEMI patients. The area under the curve (AUC) for SIRI was 0.789. The AUC for NLR was 0.754. Regarding the prediction of STEMI in different gender groups, the AUC for SIRI in the male group was 0.771. The AUC for SIRI in the female group was 0.807. Spearman correlation analysis showed that SIRI exhibited a stronger correlation with GS, while NLR was lower (SIRI: r = 0.350, P < 0.001) (NLR: r = 0.313, P < 0.001). CONCLUSION The study reveals a strong correlation between the SIRI and STEMI as well as the degree of coronary artery stenosis. In comparison to NLR, SIRI shows potential in predicting acute myocardial infarction and the severity of coronary artery stenosis. Additionally, SIRI exhibits a stronger predictive capability for female STEMI patients compared to males.
Collapse
Affiliation(s)
- Jiongchao Guo
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Yating Huang
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Lamei Pang
- Department of Endocrinology, Hefei BOE Hospital, Hefei, 230000, Anhui, China
| | - Yuan Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Jingjing Yuan
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Bingfeng Zhou
- Department of Cardiology, Hefei BOE Hospital, Hefei, 230000, Anhui, China.
| | - Minmin Fu
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China.
| |
Collapse
|
10
|
Pei G, Liu R, Wang L, He C, Fu C, Wei Q. Monocyte to high-density lipoprotein ratio is associated with mortality in patients with coronary artery diseases. BMC Cardiovasc Disord 2023; 23:451. [PMID: 37697241 PMCID: PMC10496218 DOI: 10.1186/s12872-023-03461-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/19/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Whether the monocyte to high-density lipoprotein ratio (MHR) is associated with the prognosis of coronary artery disease (CAD) is inconclusive. METHODS Patients with CAD were enrolled and their data were collected. Blood was sampled within 24 h after admission. Multivariate Cox regression analysis was performed to determine the relationship between the MHR and all-cause mortality as well as complications during hospitalization. RESULTS We included 5371 patients in our cohort study. Among them, 114 (2.12%) patients died in hospital. MHR was independently associated with all-cause mortality (hazard ratio [HR], 1.81; 95% confidence interval [CI] 1.35, 2.42), cardiovascular mortality (1.69; 1.17, 2.45) and non-cardiovascular mortality (2.04; 1.27, 3.28). This association was only observed in patients with hypertension (P for interaction = 0.003). Patients with higher MHR levels also have a higher risk of complications, including infection, pneumonia, electrolyte disturbance, gastrointestinal bleeding, multiple organ dysfunction syndrome, and disturbance of consciousness. The receiver operating characteristic (ROC) analysis showed that the MHR had higher prognostic values than monocytes and high-density lipoprotein. CONCLUSION MHR was an independent predictor of all-cause mortality and in-hospital complications in patients with CAD, especially in patients with hypertension.
Collapse
Affiliation(s)
- Gaiqin Pei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Rui Liu
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Lu Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chengqi He
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chenying Fu
- West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, Sichuan, China.
- Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Quan Wei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Wang L, Yang L, Li T, Geng S. Development and Validation of Nomogram for the Prediction of Malignant Ventricular Arrhythmia Including Circulating Inflammatory Cells in Patients with Acute ST-Segment Elevation Myocardial Infarction. J Inflamm Res 2023; 16:3185-3196. [PMID: 37529768 PMCID: PMC10389081 DOI: 10.2147/jir.s420305] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Background Malignant ventricular arrhythmia (MVA) can seriously affect the hemodynamic changes of the body. In this study, we developed and validated a nomogram to predict the in-hospital MVA risk in patients with STEMI after emergency PCI. Methods The multivariable logistic regression analysis included variables with a P<0.05 in the univariate logistic regression analysis and investigated the independent predictors affecting in-hospital MVA after PCI in patients with STEMI in the training cohort. The construction of a nomogram model used independent predictors to predict the risk of in-hospital MVA, and C-index, Hosmer-Lemeshow (HL) test, calibration curves, decision curve analysis (DCA), and receiver operating characteristic (ROC) were used to validate the nomogram. Results Killip class [OR=5.034 (95% CI: 1.596-15.809), P=0.005], CK-MB [OR=1.002 (95% CI: 1.001-1.004), P=0.022], serum potassium [OR=0.618 (95% CI: 0.406-0.918), P=0.020], NLR [OR=1.073 (95% CI: 1.034-1.115), P<0.001], and monocyte [OR=1.974 (95% CI: 1.376-2.925), P<0.001] were the independent predictors of in-hospital MVA after PCI in patients with STEMI. A nomogram including the 5 independent predictors was developed to predict the risk of in-hospital MVA. The C-index, equivalent to the area under the ROC curve (AUC), was 0.803 (95% confidence interval [CI]: 0.738-0.868) in the training cohort, and 0.801 (95% CI:0.692-0.911) in the validation cohort, showing that the nomogram had a good discrimination. The HL test (χ2=8.439, P=0.392 in the training cohort; χ2=9.730, P=0.285 in the validation cohort) revealed a good calibration. The DCA suggested an obvious clinical net benefit. Conclusion Killip class, CK-MB, serum potassium, NLR, and monocyte were independent factors for in-hospital MVA after PCI in patients with STEMI. The nomogram model constructed based on the above factors to predict the risk of in-hospital MVA had satisfactory discrimination, calibration, and clinical effectiveness, and was an excellent tool for early prediction of the risk of in-hospital MVA after PCI in patients with STEMI.
Collapse
Affiliation(s)
- Liang Wang
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, Shuyang, Jiangsu, People’s Republic of China
| | - Liting Yang
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, Shuyang, Jiangsu, People’s Republic of China
| | - Tao Li
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, Shuyang, Jiangsu, People’s Republic of China
| | - Shanshan Geng
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, Shuyang, Jiangsu, People’s Republic of China
| |
Collapse
|
12
|
Jin X, Wang X, Sun J, Tan W, Zhang G, Han J, Xie M, Zhou L, Yu Z, Xu T, Wang C, Wang Y, Zhou X, Jiang H. Subthreshold splenic nerve stimulation prevents myocardial Ischemia-Reperfusion injury via neuroimmunomodulation of proinflammatory factor levels. Int Immunopharmacol 2023; 114:109522. [PMID: 36502595 DOI: 10.1016/j.intimp.2022.109522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Clinical outcomes following myocardial ischemia-reperfusion (I/R) injury are strongly related to the intensity and duration of inflammation. The splenic nerve (SpN) is indispensable for the anti-inflammatory reflex. This study aimed to investigate whether splenic nerve stimulation (SpNS) plays a cardioprotective role in myocardial I/R injury and the potential underlying mechanism. METHODS Sprague-Dawley rats were randomly divided into four groups: sham group, I/R group, SpNS group, and I/R plus SpNS group. The highest SpNS intensity that did not influence heart rate was identified, and SpNS at this intensity was used as the subthreshold stimulus. Continuous subthreshold SpNS was applied for 1 h before ligation of the left coronary artery for 45 min. After 72 h of reperfusion, samples were collected for analysis. RESULTS SpN activity and splenic concentrations of cholinergic anti-inflammatory pathway (CAP)-related neurotransmitters were significantly increased by SpNS. The infarct size, oxidative stress, sympathetic tone, and the levels of proinflammatory cytokines, including TNF-α, IL-1β, and IL-6, were significantly reduced in rats subjected to subthreshold SpNS after myocardial I/R injury compared with those subjected to I/R injury alone. CONCLUSIONS Subthreshold SpNS ameliorates myocardial damage, the inflammatory response, and cardiac remodelling induced by myocardial I/R injury via neuroimmunomodulation of proinflammatory factor levels. SpNS is a potential therapeutic strategy for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Xiaoxing Jin
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Xiaofei Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Guocheng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Jiapeng Han
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Mengjie Xie
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Zhiyao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Tianyou Xu
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Changyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University Hubei Key Laboratory of Autonomic Nervous System Modulation Cardiac Autonomic Nervous System Research Center of Wuhan University Taikang Center for Life and Medical Sciences, Wuhan University Institute of Molecular Medicine, Renmin Hospital of Wuhan University Cardiovascular Research Institute, Wuhan University Hubei Key Laboratory of Cardiology, Wuhan 430060, P.R. China.
| |
Collapse
|
13
|
Kalinin RE, Korotkova NV, Suchkov IA, Mzhavanadze ND, Ryabkov AN. Selectins and their involvement in the pathogenesis of cardiovascular diseases. KAZAN MEDICAL JOURNAL 2022; 103:617-627. [DOI: 10.17816/kmj2022-617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The review presents current data on the structure and functional role of cell adhesion molecules belonging to the selectin family (selectins P, L and E), and their involvement in the pathogenesis of cardiovascular diseases. On the one hand, intercellular adhesion molecules of the vascular wall endothelium, platelets and leukocytes are an important link in the processes of vasculogenesis, development and regeneration of the vascular system. On the other hand, these molecules participate in the earliest stages of endothelial dysfunction with the subsequent development of pathology. For this reason, figuring out the mechanisms of activity of this group of molecules is very important for understanding the molecular basis of the cardiovascular diseases pathogenesis. The adhesion of molecules, both between cells and between cells and a component of the extracellular matrix, is the most important stage of physiological and biochemical processes. According to present knowledge, five classes of intercellular adhesion molecules are known: integrins, cadherins, immunoglobulins (including nectins), selectins and addressins. All of them are bonded to a cytoplasmic membrane and provide the interaction of cells with each other. Some of them are transmembrane and associated with the cytoskeleton of the cell. On the cell surface, intercellular adhesion molecules can be located in clusters, forming multipoint binding sites and thereby determining the degree of avidity. One of the most significant functions of selectins is participation in the initial stage of the leukocyte adhesion cascade, which results in their binding to the endothelium, rolling and further extravasation into tissues. The first stage of this process is mediated by specific non-covalent interactions between selectins and their glycan ligands, with the glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains one of the main strategies aimed at developing new methods of treating immune, inflammatory and oncological diseases.
Collapse
|
14
|
Kaldirim M, Lang A, Pfeiler S, Fiegenbaum P, Kelm M, Bönner F, Gerdes N. Modulation of mTOR Signaling in Cardiovascular Disease to Target Acute and Chronic Inflammation. Front Cardiovasc Med 2022; 9:907348. [PMID: 35845058 PMCID: PMC9280721 DOI: 10.3389/fcvm.2022.907348] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023] Open
Abstract
Inflammation is a key component in the pathogenesis of cardiovascular diseases causing a significant burden of morbidity and mortality worldwide. Recent research shows that mammalian target of rapamycin (mTOR) signaling plays an important role in the general and inflammation-driven mechanisms that underpin cardiovascular disease. mTOR kinase acts prominently in signaling pathways that govern essential cellular activities including growth, proliferation, motility, energy consumption, and survival. Since the development of drugs targeting mTOR, there is proven efficacy in terms of survival benefit in cancer and allograft rejection. This review presents current information and concepts of mTOR activity in myocardial infarction and atherosclerosis, two important instances of cardiovascular illness involving acute and chronic inflammation. In experimental models, inhibition of mTOR signaling reduces myocardial infarct size, enhances functional remodeling, and lowers the overall burden of atheroma. Aside from the well-known effects of mTOR inhibition, which are suppression of growth and general metabolic activity, mTOR also impacts on specific leukocyte subpopulations and inflammatory processes. Inflammatory cell abundance is decreased due to lower migratory capacity, decreased production of chemoattractants and cytokines, and attenuated proliferation. In contrast to the generally suppressed growth signals, anti-inflammatory cell types such as regulatory T cells and reparative macrophages are enriched and activated, promoting resolution of inflammation and tissue regeneration. Nonetheless, given its involvement in the control of major cellular pathways and the maintenance of a functional immune response, modification of this system necessitates a balanced and time-limited approach. Overall, this review will focus on the advancements, prospects, and limits of regulating mTOR signaling in cardiovascular disease.
Collapse
Affiliation(s)
- Madlen Kaldirim
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander Lang
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Pia Fiegenbaum
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital, Heinrich-Heine University, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
15
|
Zhao Y, Hao C, Bo X, Lu Z, Qian H, Chen L. The prognostic value of admission lymphocyte-to-monocyte ratio in critically ill patients with acute myocardial infarction. BMC Cardiovasc Disord 2022; 22:308. [PMID: 35799102 PMCID: PMC9264617 DOI: 10.1186/s12872-022-02745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/30/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Inflammation plays a critical role in acute myocardial infarction (AMI). Recent studies have shown the value of hematologic indicators in MI risk stratification and prognostic assessment. However, the association between lymphocyte-to-monocyte ratio (LMR) and the long-term mortality of critically ill MI patients remains unclear. METHODS Clinical data were extracted from the Medical Information Mart for Intensive Care III database. Patients diagnosed with AMI on admission in the intensive care units were include. The optimal cutoff value of LMR was determined by X-tile software. The Cox proportional hazard model was applied for the identification of independent prognostic factors of 1-year mortality and survival curves were estimated using the Kaplan-Meier method. In order to reduce selection bias, a 1:1 propensity score matching (PSM) method was performed. RESULTS A total of 1517 AMI patients were included in this study. The cutoff value for 1-year mortality of LMR determined by X-Tile software was 3.00. A total of 534 pairs of patients were matched after PSM. Multivariate analysis (HR = 1.369, 95%CI 1.110-1.687, P = 0.003) and PSM subgroups (HR = 1.299, 95%CI 1.032-1.634, P = 0.026) showed that 1-year mortality was significantly higher in patients with LMR < 3.00 than patients with LMR ≥ 3.00 in Cox proportional hazard models. The survival curves showed that patients with LMR < 3.00 had a significantly lower 1-year survival rate before (63.83 vs. 81.03%, Log rank P < 0.001) and after PSM (68.13 vs. 74.22%, Log rank P = 0.041). CONCLUSION In this retrospective cohort analysis, we demonstrated that a low admission LMR (< 3.00) was associated with a higher risk of 1-year mortality in critically ill patients with AMI.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Xiangwei Bo
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Zhengri Lu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Hao Qian
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China.
- School of Medicine, Southeast University, Nanjing, China.
- Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China.
| |
Collapse
|
16
|
Bromage DI, Trevelin SC, Huntington J, Yang VX, Muthukumar A, Mackie SJ, Sawyer G, Zhang X, Santos CXC, Safinia N, Smyrnias I, Giacca M, Ivetic A, Shah AM. Nrf2 attenuates the innate immune response after experimental myocardial infarction. Biochem Biophys Res Commun 2022; 606:10-16. [PMID: 35338853 DOI: 10.1016/j.bbrc.2022.03.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND There is compelling evidence implicating dysregulated inflammation in the mechanism of ventricular remodeling and heart failure (HF) after MI. The transcription factor nuclear factor erythroid-derived 2-like 2 (Nrf2, encoded by Nfe2l2) is a promising target in this context since it impedes transcriptional upregulation of pro-inflammatory cytokines and is anti-inflammatory in various murine models. OBJECTIVES We aimed to investigate the contribution of Nrf2 to the inflammatory response after experimental myocardial infarction (MI). METHODS We subjected Nrf2-/- mice and wild type (WT) controls to permanent left coronary artery (LCA) ligation. The inflammatory response was investigated with fluorescence-activated cell sorting (FACS) analysis of peripheral blood and heart cell suspensions, together with qRT-PCR of infarcted tissue for chemokines and their receptors. To investigate whether Nrf2-mediated transcription is a dedicated function of leukocytes, we interrogated publicly available RNA-sequencing (RNA-seq) data from mouse hearts after permanent LCA ligation for Nrf2-regulated gene (NRG) expression. RESULTS FACS analysis demonstrated a profoundly inflamed phenotype in the hearts of global Nrf2-/- mice as compared to WT mice after MI. Moreover, infarcted tissue from Nrf2-/- mice displayed higher expression of mRNA coding for inflammatory cytokines, chemokines, and their receptors, including IL-6, Ccl2, and Cxcr4. RNA-seq analysis showed upregulated NRG expression in WT mice after MI compared to naive mice, which was significantly higher in bioinformatically isolated CCR2+ cells. CONCLUSIONS Taken together, the results suggest that Nrf2 signalling in leukocytes, and possibly CCR2+ monocytes and monocyte-derived cardiac resident macrophages, may be potential targets to prevent post-MI ventricular remodeling.
Collapse
Affiliation(s)
- Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| | - Silvia C Trevelin
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Josef Huntington
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Victoria X Yang
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Ananya Muthukumar
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Sarah J Mackie
- School of Cancer and Pharmaceutical Sciences, SGDP Centre, King's College London, Memory Lane, London, SE5 8AF, UK
| | - Greta Sawyer
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Xiaohong Zhang
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Niloufar Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Ioannis Smyrnias
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, GU2 7AL, UK
| | - Mauro Giacca
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| |
Collapse
|
17
|
Peterson EA, Sun J, Wang J. Leukocyte-Mediated Cardiac Repair after Myocardial Infarction in Non-Regenerative vs. Regenerative Systems. J Cardiovasc Dev Dis 2022; 9:63. [PMID: 35200716 PMCID: PMC8877434 DOI: 10.3390/jcdd9020063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Innate and adaptive leukocytes rapidly mobilize to ischemic tissues after myocardial infarction in response to damage signals released from necrotic cells. Leukocytes play important roles in cardiac repair and regeneration such as inflammation initiation and resolution; the removal of dead cells and debris; the deposition of the extracellular matrix and granulation tissue; supporting angiogenesis and cardiomyocyte proliferation; and fibrotic scar generation and resolution. By organizing and comparing the present knowledge of leukocyte recruitment and function after cardiac injury in non-regenerative to regenerative systems, we propose that the leukocyte response to cardiac injury differs in non-regenerative adult mammals such as humans and mice in comparison to cardiac regenerative models such as neonatal mice and adult zebrafish. Specifically, extensive neutrophil, macrophage, and T-cell persistence contributes to a lengthy inflammatory period in non-regenerative systems for adverse cardiac remodeling and heart failure development, whereas their quick removal supports inflammation resolution in regenerative systems for new contractile tissue formation and coronary revascularization. Surprisingly, other leukocytes have not been examined in regenerative model systems. With this review, we aim to encourage the development of improved immune cell markers and tools in cardiac regenerative models for the identification of new immune targets in non-regenerative systems to develop new therapies.
Collapse
Affiliation(s)
| | | | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (E.A.P.); (J.S.)
| |
Collapse
|
18
|
Jia F, Chen L, Fang L, Chen W. IRAK-M deletion aggravates acute inflammatory response and mitochondrial respiratory dysfunction following myocardial infarction: A bioinformatics analysis. J Proteomics 2022; 257:104512. [DOI: 10.1016/j.jprot.2022.104512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
|
19
|
Monocyte/Lymphocyte Ratio and MCHC as Predictors of Collateral Carotid Artery Disease-Preliminary Report. J Pers Med 2021; 11:jpm11121266. [PMID: 34945738 PMCID: PMC8707882 DOI: 10.3390/jpm11121266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 01/20/2023] Open
Abstract
Background: Carotid artery disease accounts for 30% of ischemic strokes in the general population. Numerous biomarkers have been investigated for predicting either the progression or the severity of the disease. The aim of this retrospective study was to compare hematologic indices among patients referred for surgical interventions due to severe carotid disease. Methods: In total, 135 patients (87 (64.4%) men and 48 (35.6%) women) with a mean age of 70 ± 8 years who underwent surgical carotid intervention were enrolled into the study. Results: A Mann–Whitney test for independent samples revealed significant differences in monocyte to lymphocyte ratio (MLR) and mean corpuscular hemoglobin concentration (MCHC) between patients with one and two (collateral) carotid diseases. The cut-off value for MLR was 0.3 (AUC = 0.654, p = 0.048, 70.0% sensitivity and 74.6% specificity) and for MHCH was 21.6. (AUC = 0.730, p < 0.001, 70.0% sensitivity and 77.2% specificity). A multivariable model of logistic regression revealed two significant parameters for collateral carotid stenosis disease including MLR > 0.3 (OR 6.19 with 95% CI 2.02–19.01, p = 0.001) and MCHC > 21.6 (OR 7.76, 95% CI 2.54–23.72, p < 0.001). Conclusions: MLR above 0.3 and MCHC above 21.6 have predictive values for colleterial carotid stenosis and may be used as easily accessible indicators for atherosclerosis severity.
Collapse
|
20
|
Effects of remote ischemic preconditioning (RIPC) and chronic remote ischemic preconditioning (cRIPC) on levels of plasma cytokines, cell surface characteristics of monocytes and in-vitro angiogenesis: a pilot study. Basic Res Cardiol 2021; 116:60. [PMID: 34651218 PMCID: PMC8516789 DOI: 10.1007/s00395-021-00901-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
Remote ischemic preconditioning (RIPC) protects the heart against myocardial ischemia/reperfusion (I/R) injury and recent work also suggested chronic remote ischemic conditioning (cRIPC) for cardiovascular protection. Based on current knowledge that systemic immunomodulatory effects of RIPC and the anti-inflammatory capacity of monocytes might be involved in cardiovascular protection, the aim of our study was to evaluate whether RIPC/cRIPC blood plasma is able to induce in-vitro angiogenesis, identify responsible factors and evaluate the effects of RIPC/cRIPC on cell surface characteristics of circulating monocytes. Eleven healthy volunteers were subjected to RIPC/cRIPC using a blood pressure cuff inflated to > 200 mmHg for 3 × 5 min on the upper arm. Plasma and peripheral blood monocytes were isolated before RIPC (Control), after 1 × RIPC (RIPC) and at the end of 1 week of daily RIPC (cRIPC) treatment. Plasma concentrations of potentially pro-angiogenic humoral factors (CXCL5, Growth hormone, IGFBP3, IL-1α, IL-6, Angiopoietin 2, VEGF, PECAM-1, sTie-2, IL-8, MCSF) were measured using custom made multiplex ELISA systems. Tube formation assays for evaluation of in-vitro angiogenesis were performed with donor plasma, monocyte conditioned culture media as well as IL-1α, CXCL5 and Growth hormone. The presence of CD14, CD16, Tie-2 and CCR2 was analyzed on monocytes by flow cytometry. Employing in-vitro tube formation assays, several parameters of angiogenesis were significantly increased by cRIPC plasma (number of nodes, P < 0.05; number of master junctions, P < 0.05; number of segments, P < 0.05) but were not influenced by culture medium from RIPC/cRIPC treated monocytes. While RIPC/cRIPC treatment did not lead to significant changes of the median plasma concentrations of any of the selected potentially pro-angiogenic humoral factors, in-depth analysis of the individual subjects revealed differences in plasma levels of IL-1α, CXCL5 and Growth hormone after RIPC/cRIPC treatment in some of the volunteers. Nevertheless, the positive effects of RIPC/cRIPC plasma on in-vitro angiogenesis could not be mimicked by the addition of the respective humoral factors alone or in combination. While monocyte conditioned culture media did not affect in-vitro tube formation, flow cytometry analyses of circulating monocytes revealed a significant increase in the number of Tie-2 positive and a decrease of CCR2 positive monocytes after RIPC/cRIPC (Tie-2: cRIPC, P < 0.05; CCR2: RIPC P < 0.01). Cardiovascular protection may be mediated by RIPC and cRIPC via a regulation of plasma cytokines as well as changes in cell surface characteristics of monocytes (e.g. Tie-2). Our results suggest that a combination of humoral and cellular factors could be responsible for the RIPC/cRIPC mediated effects and that interindividual variations seem to play a considerable part in the RIPC/cRIPC associated mechanisms.
Collapse
|
21
|
Viola M, de Jager SCA, Sluijter JPG. Targeting Inflammation after Myocardial Infarction: A Therapeutic Opportunity for Extracellular Vesicles? Int J Mol Sci 2021; 22:ijms22157831. [PMID: 34360595 PMCID: PMC8346058 DOI: 10.3390/ijms22157831] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
After myocardial infarction (MI), a strong inflammatory response takes place in the heart to remove the dead tissue resulting from ischemic injury. A growing body of evidence suggests that timely resolution of this inflammatory process may aid in the prevention of adverse cardiac remodeling and heart failure post-MI. The present challenge is to find a way to stimulate this process without interfering with the reparative role of the immune system. Extracellular vesicles (EVs) are natural membrane particles that are released by cells and carry different macromolecules, including proteins and non-coding RNAs. In recent years, EVs derived from various stem and progenitor cells have been demonstrated to possess regenerative properties. They can provide cardioprotection via several mechanisms of action, including immunomodulation. In this review, we summarize the role of the innate immune system in post-MI healing. We then discuss the mechanisms by which EVs modulate cardiac inflammation in preclinical models of myocardial injury through regulation of monocyte influx and macrophage function. Finally, we provide suggestions for further optimization of EV-based therapy to improve its potential for the treatment of MI.
Collapse
Affiliation(s)
- Margarida Viola
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| |
Collapse
|
22
|
Association of lymphocyte-to-monocyte ratio with the long-term outcome after hospital discharge in patients with ST-elevation myocardial infarction: a retrospective cohort study. Coron Artery Dis 2021; 31:248-254. [PMID: 31658149 DOI: 10.1097/mca.0000000000000818] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Lymphocyte-to-monocyte ratio (LMR), a novel systemic inflammatory factor, correlates with adverse outcomes in patients with cardiovascular disease. However, data are limited regarding the prognostic value of LMR in patients with ST-elevation myocardial infarction (STEMI) after hospital discharge. Therefore, the aim of our study was to evaluate the prognostic impact of admission LMR in hospital survivors of STEMI. METHODS This retrospective observational study enrolled 1369 STEMI patients between 2014 and 2017. The study population was divided into three groups according to tertiles (T) of LMR (T1: ≥2.84; T2: 1.85-2.83; T3: <1.85). The primary outcomes were long-term outcomes after discharge including major adverse cardiac events (MACE) and all-cause mortality. The associations between LMR and long-term outcomes were assessed using Cox regression analysis. RESULTS The median follow-up period was 556 days (interquartile range, 342-864 days). Independent correlations were observed between LMR and both long-term MACE and all-cause mortality. For long-term MACE, the T3 (adjusted hazard ratio [HR], 1.74; 95% confidence interval [CI]: 1.12-2.70; P = 0.013) and T2 groups (adjusted HR, 1.65; CI: 1.07-2.54; P = 0.024) showed significantly higher risk of MACE than did the T1 group. For long-term all-cause mortality, the adjusted HR was 3.07 (CI: 1.10-8.54; P = 0.032) in the T3 group and 2.35 (CI: 0.82-6.76; P = 0.112) in the T2 group compared with that of the T1 group. CONCLUSION Decreased admission LMR was independently associated with long-term all-cause mortality and MACE after discharge in patients with STEMI.
Collapse
|
23
|
Dounousi E, Duni A, Naka KK, Vartholomatos G, Zoccali C. The Innate Immune System and Cardiovascular Disease in ESKD: Monocytes and Natural Killer Cells. Curr Vasc Pharmacol 2021; 19:63-76. [PMID: 32600233 DOI: 10.2174/1570161118666200628024027] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Adverse innate immune responses have been implicated in several disease processes, including cardiovascular disease (CVD) and chronic kidney disease (CKD). The monocyte subsets natural killer (NK) cells and natural killer T (NKT) cells are involved in innate immunity. Monocytes subsets are key in atherogenesis and the inflammatory cascade occurring in heart failure. Upregulated activity and counts of proinflammatory CD16+ monocyte subsets are associated with clinical indices of atherosclerosis, heart failure syndromes and CKD. Advanced CKD is a complex state of persistent systemic inflammation characterized by elevated expression of proinflammatory and pro-atherogenic CD14++CD16+ monocytes, which are associated with cardiovascular events and death both in the general population and among patients with CKD. Diminished NK cells and NKT cells counts and aberrant activity are observed in both coronary artery disease and end-stage kidney disease. However, evidence of the roles of NK cells and NKT cells in atherogenesis in advanced CKD is circumstantial and remains to be clarified. This review describes the available evidence regarding the roles of specific immune cell subsets in the pathogenesis of CVD in patients with CKD. Future research is expected to further uncover the links between CKD associated innate immune system dysregulation and accelerated CVD and will ideally be translated into therapeutic targets.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Anila Duni
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- 2nd Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece
| | - Carmine Zoccali
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Controversy exists whether beta-blockers should be given before primary percutaneous coronary intervention (PCI) or to defer their administration for up to 24 hours. RECENT FINDINGS Animal studies, most of them conducted in the 1970s and 1980s, showed evidence that early beta-blocker administration may reduce infarct size. Subsequent human studies had mixed results on infarct size and survival. More specifically, in the current primary PCI era, only four studies evaluated the impact of early intravenous beta-blocker administration after acute myocardial infarction, only two of them before PCI. All studies agree that in hemodynamically stable patients, early intravenous beta-blocker administration is safe and protected against malignant arrhythmias. Nevertheless, results on infarct size and mortality are equivocal. Considering the heterogeneity of currently available data, further studies are still needed to assess the benefit of early injection of metoprolol in STEMI patients in a large double-blinded and randomized design versus placebo.
Collapse
Affiliation(s)
- Georgios Giannakopoulos
- Cardiology Division, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Stephane Noble
- Cardiology Division, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
25
|
Pluijmert NJ, Atsma DE, Quax PHA. Post-ischemic Myocardial Inflammatory Response: A Complex and Dynamic Process Susceptible to Immunomodulatory Therapies. Front Cardiovasc Med 2021; 8:647785. [PMID: 33996944 PMCID: PMC8113407 DOI: 10.3389/fcvm.2021.647785] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 01/04/2023] Open
Abstract
Following acute occlusion of a coronary artery causing myocardial ischemia and implementing first-line treatment involving rapid reperfusion, a dynamic and balanced inflammatory response is initiated to repair and remove damaged cells. Paradoxically, restoration of myocardial blood flow exacerbates cell damage as a result of myocardial ischemia-reperfusion (MI-R) injury, which eventually provokes accelerated apoptosis. In the end, the infarct size still corresponds to the subsequent risk of developing heart failure. Therefore, true understanding of the mechanisms regarding MI-R injury, and its contribution to cell damage and cell death, are of the utmost importance in the search for successful therapeutic interventions to finally prevent the onset of heart failure. This review focuses on the role of innate immunity, chemokines, cytokines, and inflammatory cells in all three overlapping phases following experimental, mainly murine, MI-R injury known as the inflammatory, reparative, and maturation phase. It provides a complete state-of-the-art overview including most current research of all post-ischemic processes and phases and additionally summarizes the use of immunomodulatory therapies translated into clinical practice.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
26
|
Monguió-Tortajada M, Prat-Vidal C, Moron-Font M, Clos-Sansalvador M, Calle A, Gastelurrutia P, Cserkoova A, Morancho A, Ramírez MÁ, Rosell A, Bayes-Genis A, Gálvez-Montón C, Borràs FE, Roura S. Local administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction. Bioact Mater 2021; 6:3314-3327. [PMID: 33778207 PMCID: PMC7973387 DOI: 10.1016/j.bioactmat.2021.02.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The administration of extracellular vesicles (EV) from mesenchymal stromal cells (MSC) is a promising cell-free nanotherapy for tissue repair after myocardial infarction (MI). However, the optimal EV delivery strategy remains undetermined. Here, we designed a novel MSC-EV delivery, using 3D scaffolds engineered from decellularised cardiac tissue as a cell-free product for cardiac repair. EV from porcine cardiac adipose tissue-derived MSC (cATMSC) were purified by size exclusion chromatography (SEC), functionally analysed and loaded to scaffolds. cATMSC-EV markedly reduced polyclonal proliferation and pro-inflammatory cytokines production (IFNγ, TNFα, IL12p40) of allogeneic PBMC. Moreover, cATMSC-EV recruited outgrowth endothelial cells (OEC) and allogeneic MSC, and promoted angiogenesis. Fluorescently labelled cATMSC-EV were mixed with peptide hydrogel, and were successfully retained in decellularised scaffolds. Then, cATMSC-EV-embedded pericardial scaffolds were administered in vivo over the ischemic myocardium in a pig model of MI. Six days from implantation, the engineered scaffold efficiently integrated into the post-infarcted myocardium. cATMSC-EV were detected within the construct and MI core, and promoted an increase in vascular density and reduction in macrophage and T cell infiltration within the damaged myocardium. The confined administration of multifunctional MSC-EV within an engineered pericardial scaffold ensures local EV dosage and release, and generates a vascularised bioactive niche for cell recruitment, engraftment and modulation of short-term post-ischemic inflammation.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Prat-Vidal
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Miriam Moron-Font
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Marta Clos-Sansalvador
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Alexandra Calle
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Paloma Gastelurrutia
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Adriana Cserkoova
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Anna Morancho
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Miguel Ángel Ramírez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, UAB, Barcelona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc E Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona, 08500, Spain
| |
Collapse
|
27
|
Zhou Z, Liang M, Wu H, Huang S, Weng R, Hou J, Wu Z. Preoperative Lymphocyte-to-Monocyte Ratio as a Prognostic Predictor of Long-Term Mortality in Cardiac Surgery Patients: A Propensity Score Matching Analysis. Front Cardiovasc Med 2021; 8:639890. [PMID: 33693038 PMCID: PMC7937643 DOI: 10.3389/fcvm.2021.639890] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
Aims: To evaluate the prognostic value of the preoperative lymphocyte-to-monocyte ratio (LMR) in patients who underwent cardiac surgery. Methods: Clinical data were extracted from the Medical Information Mart for Intensive Care III (MIMIC-III) database. The optimal cutoff value of LMR was determined by X-tile software. The Cox proportional hazard model was applied for the identification of independent prognostic factors of 4-year mortality and survival curves were estimated using the Kaplan-Meier method. In order to balance the influence of potential confounding factors, a 1:1 propensity score matching (PSM) method was performed. Results: A total of 1,701 patients were included. The X-tile software indicated that the optimal cutoff value of the LMR for 4-year mortality was 3.58. After PSM, 489 pairs of score-matched patients were generated. The Cox proportional hazard model showed that patients with an LMR < 3.58 had a significantly higher 4-year mortality than patients with an LMR ≥ 3.58 in the entire cohort (HR = 1.925, 95%CI: 1.509-2.456, p < 0.001) and the PSM subset (HR = 1.568, 95%CI: 1.2-2.05, p = 0.001). The survival curves showed that patients with an LMR < 3.58 had a significant lower 4-year survival rate in the entire cohort (71.7 vs. 88.5%, p < 0.001) and the PSM subset (73.2 vs. 81.4%, p = 0.002). Conclusions: A lower LMR (<3.58) was associated with a higher risk of 4-year mortality and can serve as a prognostic predictor of the long-term mortality in cardiac surgery patients.
Collapse
Affiliation(s)
- Zhuoming Zhou
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mengya Liang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Huawei Wu
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, United States
| | - Suiqing Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Rennan Weng
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jian Hou
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Mo F, Luo Y, Yan Y, Li J, Lai S, Wu W. Are activated B cells involved in the process of myocardial fibrosis after acute myocardial infarction? An in vivo experiment. BMC Cardiovasc Disord 2021; 21:5. [PMID: 33407160 PMCID: PMC7789158 DOI: 10.1186/s12872-020-01775-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inflammatory cells infiltrate into the ischemic and hypoxic myocardial tissue after myocardial infarction. B cells gather at the site of myocardial injury and secrete cytokines to regulate immune inflammation and fiber repair processes. METHODS The animal experiment used ligation of the left anterior descending (LAD) artery of C57BL/6 mice to establish a mouse acute myocardial infarction (AMI) model to observe changes in activated B cells and cytokines at different time points. Twelve-week-old C57BL/6 male mice were randomly divided into the Sham group (24 mice) (thread under the LAD artery without ligation) and the AMI group (64 mice). In addition, C57BL/6 B-cell knockout (BKO) mice and C57BL/6 wild-type (WT) mice were used to establish AMI models to observe the expression levels of cardiomyocyte cytokines, such as TNF-α IL-1β, IL-6, TGF-β1, COL1-A1, COL3-AIII, TIMP, and MMP9. Moreover, pathological and collagen changes in the myocardium were analysed. One-way ANOVA and LSD method was used for comparisons of multiple and pairwise groups respectively. P < 0.05 indicated significant differences. RESULTS An AMI model of C57BL/6 mice was established successfully. The ratio of activated B cells and the expression of TNF-α, IL-1β, IL-6, TGF-β1, and B cell activating factor (BAFF) in the 5-day subgroup were the highest in the myocardium, spleen and peripheral blood with the most obvious myocardial inflammatory cell infiltration. The cytokines mRNA expression levels in the 5-day subgroup of the BKO group were decreased compared with those in the WT group (P < 0.05). Among the 2-week subgroups of the Sham, WT and BKO groups, the the LVEDd and LVESd of the BKO group were lower than those of the WT group (P < 0.05), and the left ventricular ejection fraction was higher than that of the WT group (P < 0.05). CONCLUSION Activated B cells participate in the sustained state of myocardial inflammation and immune system activation after AMI, and may affect the metabolism of myocardial collagen after AMI by secreting cytokines. Moreover, B cells promote the expression of myocardial collagen Type I and Type III and damage the left ventricular ejection function.
Collapse
Affiliation(s)
- Fanrui Mo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
- Department of Cardiology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Ying Luo
- Guangxi Medical University, Nanning, China
| | - Yuluan Yan
- Department of Cardiology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Juan Li
- Department of Cardiology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shayi Lai
- Department of Cardiology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Weifeng Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
29
|
Si Y, Liu J, Shan W, Zhang Y, Han C, Wang R, Sun L. Association of lymphocyte-to-monocyte ratio with total coronary plaque burden in patients with coronary artery disease. Coron Artery Dis 2020; 31:650-655. [PMID: 32097130 PMCID: PMC7531493 DOI: 10.1097/mca.0000000000000857] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/19/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Lymphocyte-to-monocyte ratio (LMR) is involved in all stages of coronary atherosclerosis and related to coronary artery disease (CAD). However, the correlation between LMR and the coronary plaque burden of CAD is not clearly elucidated. Therefore, this study aimed to investigate their correlation in patients with CAD. METHODS A total of 1953 consecutive eligible inpatients with suspected CAD were retrospectively included in this study. They were assigned into CAD (n = 564) and non-CAD groups (n = 1389). All patients underwent coronary computed tomographic angiography to evaluate coronary stenosis and coronary artery calcification (CAC). Spearman's tests were used to analyze the correlation between CAC score and LMR. Multivariate logistic regression models were set up to assess the risk factors of CAD. RESULTS Patients with CAD had lower LMR value than patients without CAD (P = 0.001). LMR was negatively correlated with CAC score and was an independent risk factor of CAC score (P < 0.05). Multivariate logistic regression model showed that LMR ≤4.8 was a newly independent risk factor of CAD (all P < 0.05). Additionally, the new risk score model was compared with the Framingham model and showed that NRI was 4.9%, which proved that the new risk score model improved the prediction capability of CAD. CONCLUSION LMR ≤4.8 is a new independent risk factor of CAD. LMR value was negatively correlated with CAC score and could be used as a new marker to evaluate the coronary plaque burden of CAD.
Collapse
Affiliation(s)
- Yueqiao Si
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Jingyi Liu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Weichao Shan
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Ying Zhang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Chao Han
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Ruijuan Wang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, HeBei, China
| |
Collapse
|
30
|
Lu S, Tian Y, Luo Y, Xu X, Ge W, Sun G, Sun X. Iminostilbene, a novel small-molecule modulator of PKM2, suppresses macrophage inflammation in myocardial ischemia-reperfusion injury. J Adv Res 2020; 29:83-94. [PMID: 33842007 PMCID: PMC8020153 DOI: 10.1016/j.jare.2020.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction Inflammation is a key factor in myocardial ischemia/reperfusion (MI/R) injury. Targeting leucocyte-mediated inflammation is an important strategy for MI/R therapy. Iminostilbene (ISB), a simple dibenzoazepine small molecule compound, has a strong anti-neurodegenerative effect. However, no study has shown the cardioprotective effect of ISB. Objectives This study aimed to investigate the role of ISB against MI/R injury and identify its molecular target. Methods To verify the cardiac protection of ISB in vivo and in vitro, we performed rat MI/R surgery and subjected inflammatory modeling of macrophages. In terms of molecular mechanisms, we designed and synthesized a small molecular probe of ISB and employed it on the click chemistry-activity-based protein profiling technique to fish for ISB targets in macrophages. To identify the target, we applied the competitive inhibition assay, surface-plasmon resonance (SPR), cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS) assay. Results In vivo, ISB showed robust anti-myocardial injury activity by improving cardiac function, reducing myocardial infarction, and inhibiting macrophage-mediated inflammation. In vitro, ISB strongly inhibited the transcription and the expression levels of inflammatory cytokines in macrophages. The pyruvate kinase isozyme type M2 (PKM2) was identified as the potential target of ISB through proteomic analysis and the competitive assay was performed for specific binding verification. Further thermodynamic and kinetic experiments showed that ISB was bound to PKM2 in a dose-dependent manner. Moreover, in terms of the biological function of ISB on PKM2, ISB reduced the expression of PKM2, thereby reducing the expression of HIF1α and the phosphorylation of STAT3. Conclusion This study for the first time demonstrated that ISB targeted PKM2 to reduce macrophage inflammation thereby significantly alleviating MI/R injury.
Collapse
Affiliation(s)
- Shan Lu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| | - Xudong Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| | - Wenxiu Ge
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, Heilongjiang, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine, China
- Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, China
| |
Collapse
|
31
|
Abstract
Cardiac PET/MR imaging is an integrated imaging approach that requires less radiation than PET/computed tomography and combines the high spatial resolution and morphologic data from MR imaging with the physiologic information from PET. This hybrid approach has the potential to improve the diagnostic and prognostic evaluation of several cardiovascular conditions, such as ischemic heart disease, infiltrative diseases such as sarcoidosis, acute and chronic myocarditis, and cardiac masses. Herein, the authors discuss the strengths of PET and MR imaging in several cardiovascular conditions; the challenges and potential; and the current data on the application of this powerful hybrid imaging modality.
Collapse
Affiliation(s)
- Rhanderson Cardoso
- Division of Cardiology, Johns Hopkins Hospital, 600 North Wolfe Street, Blalock 547, Baltimore, MD 21287, USA
| | - Thorsten M Leucker
- Division of Cardiology, Johns Hopkins Hospital, 600 North Wolfe Street, Blalock 547, Baltimore, MD 21287, USA.
| |
Collapse
|
32
|
Peet C, Ivetic A, Bromage DI, Shah AM. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc Res 2020; 116:1101-1112. [PMID: 31841135 PMCID: PMC7177720 DOI: 10.1093/cvr/cvz336] [Citation(s) in RCA: 285] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/03/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022] Open
Abstract
Improvements in early interventions after acute myocardial infarction (AMI), notably, the increased use of timely reperfusion therapy, have increased survival dramatically in recent decades. Despite this, maladaptive ventricular remodelling and subsequent heart failure (HF) following AMI remain a significant clinical challenge, particularly because several pre-clinical strategies to attenuate remodelling have failed to translate into clinical practice. Monocytes and macrophages, pleiotropic cells of the innate immune system, are integral in both the initial inflammatory response to injury and subsequent wound healing in many tissues, including the heart. However, maladaptive immune cell behaviour contributes to ventricular remodelling in mouse models, prompting experimental efforts to modulate the immune response to prevent the development of HF. Seminal work in macrophage biology defined macrophages as monocyte-derived cells that are comprised of two populations, pro-inflammatory M1 macrophages and reparative M2 macrophages, and initial investigations into cardiac macrophage populations following AMI suggested they aligned well to this model. However, more recent data, in the heart and other tissues, demonstrate remarkable heterogeneity and plasticity in macrophage development, phenotype, and function. These recent insights into macrophage biology may explain the failure of non-specific immunosuppressive strategies and offer novel opportunities for therapeutic targeting to prevent HF following AMI. Here, we summarize the traditional monocyte-macrophage paradigm, experimental evidence for the significance of these cells in HF after AMI, and the potential relevance of emerging evidence that refutes canonical models of monocyte and macrophage biology.
Collapse
Affiliation(s)
- Claire Peet
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
33
|
Liu Y, Xu J, Wu M, Kang L, Xu B. The effector cells and cellular mediators of immune system involved in cardiac inflammation and fibrosis after myocardial infarction. J Cell Physiol 2020; 235:8996-9004. [PMID: 32352172 DOI: 10.1002/jcp.29732] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/11/2020] [Accepted: 04/15/2020] [Indexed: 01/05/2023]
Abstract
The cardiac repair after myocardial infarction (MI) involves two phases, namely, inflammatory response and proliferative response. The former is an inflammatory reaction, evoked by different kinds of pro-inflammatory leukocytes and molecules stimulated by myocardial necrosis, while the latter is a repair process, predominated by a magnitude of anti-inflammatory cells and cytokines, as well as fibroblasts. Cardiac remodeling post-MI is dependent on the balance of individualized intensity of the post-MI inflammation and subsequent cardiac fibrosis. During the past 30 years, enormous studies have focused on investigating immune cells and mediators involved in cardiac inflammation and fibrosis, which are two interacting processes of post-MI cardiac repair. These results contribute to revealing the mechanism of adverse cardiac remodeling after MI and alleviating the impairment of cardiac function. In this study, we will broadly discuss the role of immune cell subpopulation and the involved cytokines and chemokines during cardiac repair post-MI, particular in cardiac inflammation and fibrosis.
Collapse
Affiliation(s)
- Yihai Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Jiamin Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Mingyue Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical college of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
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
|
35
|
Konijnenberg LSF, Damman P, Duncker DJ, Kloner RA, Nijveldt R, van Geuns RJM, Berry C, Riksen NP, Escaned J, van Royen N. Pathophysiology and diagnosis of coronary microvascular dysfunction in ST-elevation myocardial infarction. Cardiovasc Res 2020; 116:787-805. [PMID: 31710673 PMCID: PMC7061278 DOI: 10.1093/cvr/cvz301] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/13/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Early mechanical reperfusion of the epicardial coronary artery by primary percutaneous coronary intervention (PCI) is the guideline-recommended treatment for ST-elevation myocardial infarction (STEMI). Successful restoration of epicardial coronary blood flow can be achieved in over 95% of PCI procedures. However, despite angiographically complete epicardial coronary artery patency, in about half of the patients perfusion to the distal coronary microvasculature is not fully restored, which is associated with increased morbidity and mortality. The exact pathophysiological mechanism of post-ischaemic coronary microvascular dysfunction (CMD) is still debated. Therefore, the current review discusses invasive and non-invasive techniques for the diagnosis and quantification of CMD in STEMI in the clinical setting as well as results from experimental in vitro and in vivo models focusing on ischaemic-, reperfusion-, and inflammatory damage to the coronary microvascular endothelial cells. Finally, we discuss future opportunities to prevent or treat CMD in STEMI patients.
Collapse
Affiliation(s)
- Lara S F Konijnenberg
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Dirk J Duncker
- Department of Radiology and Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Colin Berry
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
- British Heart Foundation, Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Javier Escaned
- Department of Cardiology, Hospital Clínico San Carlos IDISSC, Universidad Complutense de Madrid, Madrid, Spain
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
36
|
Figueroa-Vega N, Marín-Aragón CI, López-Aguilar I, Ibarra-Reynoso L, Pérez-Luque E, Malacara JM. Analysis of the percentages of monocyte subsets and ILC2s, their relationships with metabolic variables and response to hypocaloric restriction in obesity. PLoS One 2020; 15:e0228637. [PMID: 32074122 PMCID: PMC7029876 DOI: 10.1371/journal.pone.0228637] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/21/2020] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Obesity results from excess energy intake over expenditure and is characterized by chronic low-grade inflammation involving circulating monocytes (Mo) and group 2 innate lymphoid cells (ILC2s) imbalance. We analyzed circulating Mo subsets and ILC2s percentages and β2-adrenergic receptor (β2AR) expression in lean and obese subjects, and the possible effect of hypocaloric restriction on these innate immune cells. METHODS In 139 individuals aged 45 to 57 years, classified in 74 lean individuals (>18.9kg/m2 BMI <24.9kg/m2) and 65 with obesity (n = 65), we collected fasting blood samples to detect Mo subsets, ILC2s number, and β2AR expression by flow cytometry. Lipids, insulin, leptin, and acylated-ghrelin concentrations were quantified. Resting energy expenditure (REE) was estimated by indirect calorimetry. These measurements were repeated in obese subjects after 7-weeks of hypocaloric restriction. RESULTS Non-classical monocytes (NCM) and β2AR expression on intermediate Mo (IM) were increased in obese individuals (p<0.001, in both cases), whereas the percent of ILC2s was decreased (p<0.0001). Stepwise regression analysis showed significantly negative associations of ILC2s with caloric intake, β2AR expression on IM with REE, but a positive relationship between NCM and HOMA-IR. Caloric restriction allowed a significant diminution of NCM and the β2AR expression on IM, as well as, an increase in the percent of classical Mo (CM), and ILC2s. ΔREE was related to ΔCD16+/CD16- ratio. CONCLUSIONS These findings show that in obesity occur changes in NCM, ILC2s and β2AR expression, which contribute to the low-grade inflammation linked to obesity and might revert with caloric restriction.
Collapse
Affiliation(s)
- Nicté Figueroa-Vega
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | | | - Itzel López-Aguilar
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Lorena Ibarra-Reynoso
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Elva Pérez-Luque
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Juan Manuel Malacara
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| |
Collapse
|
37
|
Bosch X, Jáuregui B, Villamor N, Morales-Ruiz M, Ortiz-Pérez JT, Borràs R, Penela D, Soto-Iglesias D, Perea RJ, Doltra A, Prat-González S, Jiménez W, Mira Á, Lasalvia L, Berruezo A. Monocyte Subsets Are Differently Associated with Infarct Size, Left Ventricular Function, and the Formation of a Potentially Arrhythmogenic Scar in Patients with Acute Myocardial Infarction. J Cardiovasc Transl Res 2019; 13:722-730. [PMID: 31833003 DOI: 10.1007/s12265-019-09944-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/28/2019] [Indexed: 11/25/2022]
Abstract
To investigate the role of classical (CLM, CD14++CD16-), intermediate (INTM, CD14++CD16+), and non-classical (Non-CLM, CD14+CD16++) monocytes in scar formation after ST-elevation myocardial infarction (STEMI), evaluated with cardiac magnetic resonance (CMR). One hundred two patients with a first STEMI had serial blood analyses after 1, 3, and 7 days. A CMR was performed at 7 days and 6 months, depicting scar core (CO), border zone (BZ), and the presence of BZ channels. CLM and INTM levels progressively decreased, correlated with the scar mass, CO, and BZ at 7 days and 6 months (p < 0.05), and inversely with left ventricular ejection fraction (LVEF, p < 0.01). Non-CLM levels gradually increased, correlated with BZ mass and the presence of BZ channels at 7 days and 6 months (p < 0.001).CLM and INTM are associated with infarct size and inversely with LVEF, whereas Non-CLM are associated with BZ mass and the presence of potentially arrhythmogenic substrate.
Collapse
Affiliation(s)
- Xavier Bosch
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Jáuregui
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Heart Institute, Teknon Medical Center, Barcelona, Spain
| | - Neus Villamor
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Hematopathology Unit, Pathology Department, Hospital Clínic, University of Barcelona, Spain, Barcelona, Spain
| | - Manuel Morales-Ruiz
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clínic, University of Barcelona, Spain, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - José T Ortiz-Pérez
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roger Borràs
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain
| | - Diego Penela
- Cardiology Department, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | - David Soto-Iglesias
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain.,Heart Institute, Teknon Medical Center, Barcelona, Spain
| | - Rosario J Perea
- Radiology Department, Hospital Clínic, University of Barcelona, Spain, Barcelona, Spain
| | - Ada Doltra
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain
| | - Susana Prat-González
- Cardiology Department, Cardiovascular Institute, Hospital Clínic, University of Barcelona, Spain, Carrer de Villarroel, 170, 08036, Barcelona, Spain
| | - Wladimiro Jiménez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clínic, University of Barcelona, Spain, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Áurea Mira
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Biochemistry and Molecular Genetics Department, Hospital Clínic, University of Barcelona, Spain, Barcelona, Spain
| | | | | |
Collapse
|
38
|
Sezer M, van Royen N, Umman B, Bugra Z, Bulluck H, Hausenloy DJ, Umman S. Coronary Microvascular Injury in Reperfused Acute Myocardial Infarction: A View From an Integrative Perspective. J Am Heart Assoc 2019; 7:e009949. [PMID: 30608201 PMCID: PMC6404180 DOI: 10.1161/jaha.118.009949] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Murat Sezer
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | | | - Berrin Umman
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Zehra Bugra
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Heerajnarain Bulluck
- 3 The Hatter Cardiovascular Institute Institute of Cardiovascular Science University College London London United Kingdom.,4 Papworth Hospital NHS Trust Cambridge United Kingdom
| | - Derek J Hausenloy
- 3 The Hatter Cardiovascular Institute Institute of Cardiovascular Science University College London London United Kingdom.,4 Papworth Hospital NHS Trust Cambridge United Kingdom.,5 National Heart Research Institute Singapore National Heart Centre Singapore Singapore.,6 Cardiovascular and Metabolic Disorders Program Duke-National University of Singapore Singapore.,7 Yong Loo Lin School of Medicine National University Singapore Singapore.,8 The National Institute of Health Research University College London Hospitals Biomedical Research Centre London United Kingdom.,9 Barts Heart Centre St Bartholomew's Hospital London United Kingdom
| | - Sabahattin Umman
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| |
Collapse
|
39
|
Bajpai G, Bredemeyer A, Li W, Zaitsev K, Koenig AL, Lokshina I, Mohan J, Ivey B, Hsiao HM, Weinheimer C, Kovacs A, Epelman S, Artyomov M, Kreisel D, Lavine KJ. Tissue Resident CCR2- and CCR2+ Cardiac Macrophages Differentially Orchestrate Monocyte Recruitment and Fate Specification Following Myocardial Injury. Circ Res 2019; 124:263-278. [PMID: 30582448 DOI: 10.1161/circresaha.118.314028] [Citation(s) in RCA: 441] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Recent advancements have brought to light the origins, complexity, and functions of tissue-resident macrophages. However, in the context of tissue injury or disease, large numbers of monocytes infiltrate the heart and are thought to contribute to adverse remodeling and heart failure pathogenesis. Little is understood about the diversity of monocytes and monocyte-derived macrophages recruited to the heart after myocardial injury, including the mechanisms that regulate monocyte recruitment and fate specification. OBJECTIVE We sought to test the hypothesis that distinct subsets of tissue-resident CCR2- (C-C chemokine receptor 2) and CCR2+ macrophages orchestrate monocyte recruitment and fate specification after myocardial injury. METHODS AND RESULTS We reveal that in numerous mouse models of cardiomyocyte cell death (permanent myocardial infarction, reperfused myocardial infarction, and diphtheria toxin cardiomyocyte ablation), there is a shift in macrophage ontogeny whereby tissue-resident macrophages are predominately replaced by infiltrating monocytes and monocyte-derived macrophages. Using syngeneic cardiac transplantation to model ischemia-reperfusion injury and distinguish tissue-resident from recruited cell populations in combination with intravital 2-photon microscopy, we demonstrate that monocyte recruitment is differentially orchestrated by distinct subsets of tissue-resident cardiac macrophages. Tissue-resident CCR2+ macrophages promote monocyte recruitment through an MYD88 (myeloid differentiation primary response 88)-dependent mechanism that results in release of MCPs (monocyte chemoattractant proteins) and monocyte mobilization. In contrast, tissue-resident CCR2- macrophages inhibit monocyte recruitment. Using CD (cluster of differentiation) 169-DTR (diphtheria toxin receptor) and CCR2-DTR mice, we further show that selective depletion of either tissue-resident CCR2- or CCR2+ macrophages before myocardial infarction results in divergent effects on left ventricular function, myocardial remodeling, and monocyte recruitment. Finally, using single-cell RNA sequencing, we show that tissue-resident cardiac macrophages differentially instruct monocyte fate specification. CONCLUSIONS Collectively, these observations establish the mechanistic basis by which monocytes are initially recruited to the injured heart and provide new insights into the heterogeneity of monocyte-derived macrophages.
Collapse
Affiliation(s)
- Geetika Bajpai
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Andrea Bredemeyer
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Wenjun Li
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Konstantin Zaitsev
- Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Andrew L Koenig
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Inessa Lokshina
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Jayaram Mohan
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Brooke Ivey
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - His-Min Hsiao
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Carla Weinheimer
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Attila Kovacs
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Slava Epelman
- Toronto General Hospital Research Institute, Division of Cardiology, University Health Network, ON, Canada (S.E.)
| | - Maxim Artyomov
- Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Daniel Kreisel
- Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO
| | - Kory J Lavine
- From the Department of Medicine (G.B., A.B., A.L. Koenig, I.L., J.M., B.I., C.W., A. Kovacs, K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Surgery (W.L., H.-M.H., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Immunology and Pathology (K.Z., M.A., D.K., K.J.L.), Washington University School of Medicine, St. Louis, MO.,Department of Developmental Biology (K.J.L.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
40
|
Mangold A, Hofbauer TM, Ondracek AS, Artner T, Scherz T, Speidl WS, Krychtiuk KA, Sadushi-Kolici R, Jakowitsch J, Lang IM. Neutrophil extracellular traps and monocyte subsets at the culprit lesion site of myocardial infarction patients. Sci Rep 2019; 9:16304. [PMID: 31704966 PMCID: PMC6841683 DOI: 10.1038/s41598-019-52671-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Neutrophils release their chromatin into the extracellular space upon activation. These web-like structures are called neutrophil extracellular traps (NETs) and have potent prothrombotic and proinflammatory properties. In ST-elevation myocardial infarction (STEMI), NETs correlate with increased infarct size. The interplay of neutrophils and monocytes impacts cardiac remodeling. Monocyte subsets are classified as classical, intermediate and non-classical monocytes. In the present study, in vitro stimulation with NETs led to an increase of intermediate monocytes and reduced expression of CX3CR1 in all subsets. Intermediate monocytes have been associated with poor outcome, while non-classical CX3CR1-positive monocytes could have reparative function after STEMI. We characterized monocyte subsets and NET markers at the culprit lesion site of STEMI patients (n = 91). NET surrogate markers were increased and correlated with larger infarct size and with fewer non-classical monocytes. Intermediate and especially non-classical monocytes were increased at the culprit site compared to the femoral site. Low CX3CR1 expression of monocytes correlated with high NET markers and increased infarct size. In this translational system, causality cannot be proven. However, our data suggest that NETs interfere with monocytic differentiation and receptor expression, presumably promoting a subset shift at the culprit lesion site. Reduced monocyte CX3CR1 expression may compromise myocardial salvage.
Collapse
Affiliation(s)
- Andreas Mangold
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Thomas M Hofbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Anna S Ondracek
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Tyler Artner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Thomas Scherz
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Walter S Speidl
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Konstantin A Krychtiuk
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Roela Sadushi-Kolici
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Johannes Jakowitsch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Irene M Lang
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, Rosenthal N, Kovacic JC. The Macrophage in Cardiac Homeostasis and Disease: JACC Macrophage in CVD Series (Part 4). J Am Coll Cardiol 2019; 72:2213-2230. [PMID: 30360829 DOI: 10.1016/j.jacc.2018.08.2149] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022]
Abstract
Macrophages are integral components of cardiac tissue and exert profound effects on the healthy and diseased heart. Paradigm shifting studies using advanced molecular techniques have revealed significant complexity within these macrophage populations that reside in the heart. In this final of a 4-part review series covering the macrophage in cardiovascular disease, the authors review the origins, dynamics, cell surface markers, and respective functions of each cardiac macrophage subset identified to date, including in the specific scenarios of myocarditis and after myocardial infarction. Looking ahead, a deeper understanding of the diverse and often dichotomous functions of cardiac macrophages will be essential for the development of targeted therapies to mitigate injury and orchestrate recovery of the diseased heart. Moreover, as macrophages are critical for cardiac healing, they are an emerging focus for therapeutic strategies aimed at minimizing cardiomyocyte death, ameliorating pathological cardiac remodeling, and for treating heart failure and after myocardial infarction.
Collapse
Affiliation(s)
- Kory J Lavine
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri; Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, Melbourne, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, Toronto, Ontario, Canada
| | - Benjamin J Kopecky
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, Maine; Mt. Desert Island Biological Laboratory, Bar Harbor, Maine
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
42
|
Charach G, Rogowski O, Karniel E, Charach L, Grosskopf I, Novikov I. Monocytes may be favorable biomarker and predictor of long-term outcome in patients with chronic heart failure: A cohort study. Medicine (Baltimore) 2019; 98:e17108. [PMID: 31567947 PMCID: PMC6756710 DOI: 10.1097/md.0000000000017108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although some studies found that an increased monocyte count is a predictive, short-term marker of unfavorable outcomes for patients with acute heart failure (HF), others have reported that monocytosis predicts prolonged survival.The current follow-up study aimed to identify different monocyte count patterns and their prognostic association with HF outcomes.Baseline blood samples for complete blood counts, differential counts, renal function tests, and lipid profiles of 303 chronic HF patients (average NYHA classification 2.8) were prospectively obtained to evaluate whether there is an association between monocyte count and clinical outcomes.Mean follow-up was 11.3 years (range 1 month to 16 years) and 111 (36.6%) patients died during follow-up. Mean monocyte count was 10.6 ± 5.5 and mean left ventricular ejection fraction (LVEF) was 36%. Patients with low monocyte counts (≤6%) had significantly lower survival rates than did those with monocyte counts 6.1% to 14%, or >14% (14.3% vs 70.2% vs. 88%, P < .001). Poorest survival was predicted for patients with NYHA class 3 to 4 and monocyte counts ≤6. Regression analysis showed that monocyte levels, NYHA class, and LVEF values were predictors of mortality, in decreasing importance.The total monocyte count was found to be an important prognostic factor that was inversely associated with predicted long-term mortality among patients with chronic HF. A low total monocyte count was strongly correlated with NYHA class and B-type natriuretic peptide levels, but no correlation was found with LVEF and oxidized low-density lipoproteins. It emerged as an independent risk factor for mortality in patients with chronic HF.
Collapse
Affiliation(s)
- Gideon Charach
- Department of Internal Medicine C, Tel Aviv Sourasky Medical Center
- Department of Internal Medicine B, Meir Medical Center, Kfar Saba, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ori Rogowski
- Department of Internal Medicine C, Tel Aviv Sourasky Medical Center
| | - Eli Karniel
- Department of Internal Medicine B, Meir Medical Center, Kfar Saba, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Charach
- Department of Internal Medicine C, Tel Aviv Sourasky Medical Center
| | - Itamar Grosskopf
- Department of Internal Medicine C, Tel Aviv Sourasky Medical Center
| | - Ilya Novikov
- Department of Internal Medicine C, Tel Aviv Sourasky Medical Center
| |
Collapse
|
43
|
Electronegative LDL from Rabbits Fed with Atherogenic Diet Is Highly Proinflammatory. Mediators Inflamm 2019; 2019:6163130. [PMID: 31534437 PMCID: PMC6724430 DOI: 10.1155/2019/6163130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL(-)) has been found in the plasma of familial hypercholesterolemia and acute myocardial infarction and has been implicated in atherosclerosis and cardiovascular disease. However, less is known about the involvement of LDL(-) in atherosclerosis-related inflammation. This study aims at investigating the inducibility of LDL(-) by atherogenic diet in rabbits and at exploring the proinflammatory potential of the diet-induced LDL(-) in macrophages. Rabbits were fed with an atherogenic diet; LDL was isolated from plasma by NaBr density gradient ultracentrifugation and was then resolved into nLDL and LDL(-) by anion-exchange chromatography. Isolated nLDL and LDL(-) were directly used or incubated with 10 μM CuSO4 for 24 h to produce copper- (Cu-) ox-nLDL and Cu-ox-LDL(-). The effects of these LDLs on inflammation were evaluated in THP-1-derived macrophages. Macrophages were treated with nLDL, LDL(-), and extensively oxidized LDL (ox-LDL), then the levels of interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α in a culture medium were determined by ELISA, and the levels of total and phosphorylated IκB, p65, p38, JNK, and ERK in cell lysates were determined by Western blotting. The LDL(-) induced significantly higher levels of IL-1β, IL-6, and TNF-α in the medium. The levels of phosphorylated/total IκB, p65, p38, JNK, and ERK were also upregulated by LDL(-). In contrast, nLDL, Cu-ox-nLDL, and Cu-ox-LDL(-) exhibited much less effect. Knockdown of lectin-type oxidized LDL receptor- (LOX-) 1 resulted in significant reduction in LDL(-)-induced IL-1β, IL-6, and TNF-α. In addition, these LDL(-) effects were also markedly attenuated by inhibition of NF-κB and ERK1/2. The data suggested that LDL(-) induced inflammation through LOX-1-, NF-κB-, and ERK1/2-dependent pathways. Taken together, our results show that rabbits fed with atherogenic diet produce a highly proinflammatory LDL(-) that is more potent in inducing inflammation than nLDL and extensively oxidize LDL in macrophages. The results thus provide a novel link between diet-induced hypercholesterolemia and inflammation.
Collapse
|
44
|
Fei YD, Wang Q, Hou JW, Li W, Cai XX, Yang YL, Zhang LH, Wei ZX, Chen TZ, Wang YP, Li YG. Macrophages facilitate post myocardial infarction arrhythmias: roles of gap junction and KCa3.1. Am J Cancer Res 2019; 9:6396-6411. [PMID: 31588225 PMCID: PMC6771231 DOI: 10.7150/thno.34801] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
Effective therapeutic targets against post-myocardial infarction (MI) arrhythmias remain to be discovered. We aimed to investigate the role of macrophages in post-MI arrhythmias. Methods: Mononuclear cell accumulation, macrophage polarization from M0 to M1 subset, and gap junction formation were analyzed in MI patients and MI mice by flow cytometry, immunofluorescence and patch clamping. Differentially expressed genes were identified by RNA sequencing. Macrophages and cardiomyocytes were cocultured in vitro, and the effects of gap junction and KCa3.1 on electrophysiological properties were assessed by patch clamping. The effects of KCa3.1 inhibition on post-MI arrhythmias were assessed by intracardiac stimulation and ambulatory electrocardiograms in vivo. Results: Percentage of pro-inflammatory mononuclear cells were significantly elevated in patients with post-MI arrhythmias compared with MI patients without arrhythmias and healthy controls (p<0.001). Macrophages formed gap junction with cardiomyocytes in MI border zones of MI patient and mice, and pro-inflammatory macrophages were significantly increased 3 days post-MI (p<0.001). RNA sequencing identified Kcnn4 as the most differentially expressed gene encoding ion channel, and the upregulation is mainly attributed to macrophage accumulation and polarization into pro-inflammatory subset. In vitro coculture experiments demonstrated that connection with M0 macrophages via gap junction slightly shortened the action potential durations (APDs) of cardiomyocytes. However, the APD90 of cardiomyocytes connected with M1 macrophages were significantly prolonged (p<0.001), which were effectively attenuated by gap junction inhibition (p=0.002), KCa3.1 inhibition (p=0.008), KCa3.1 silencing (p<0.001) and store-operated Ca2+ channel inhibition (p=0.005). In vivo results demonstrated that KCa3.1 inhibition significantly decreased the QTc durations (p=0.031), intracardiac stimulation-induced ventricular arrhythmia durations (p=0.050) and incidence of premature ventricular contractions (p=0.030) in MI mice. Conclusion: Macrophage polarization leads to APD heterogeneity and post-MI arrhythmias via gap junction and KCa3.1 activation. The results provide evidences of a novel mechanism of post-MI heterogeneous repolarization and arrhythmias, rendering macrophages and KCa3.1 to be potential therapeutic targets.
Collapse
|
45
|
Çekici Y, Yılmaz M, Seçen Ö. New inflammatory indicators: association of high eosinophil-to-lymphocyte ratio and low lymphocyte-to-monocyte ratio with smoking. J Int Med Res 2019; 47:4292-4303. [PMID: 31319727 PMCID: PMC6753567 DOI: 10.1177/0300060519862077] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective Smoking has been proven to increase systemic inflammation in previous studies
using different biomarkers. The eosinophil-to-lymphocyte ratio (ELR),
neutrophil-to-lymphocyte ratio (NLR), and lymphocyte-to-monocyte ratio (LMR)
are new indicators of systemic inflammation that are used as predictors of
systemic inflammation, morbidity, and mortality associated with many
diseases. We investigated the effects of smoking on these inflammatory
indicators. Methods In total, 616 consecutive smoking healthy subjects and 387 age-matched
nonsmoking healthy subjects were enrolled. White blood cell counts
(neutrophils, lymphocytes, basophils, eosinophils, and monocytes) were
determined by electrical impedance with an automatic blood cell counting
device. The ELR, LMR, and NLR were calculated based on these cell counts.
Smoking habits of participants were calculated as pack/year. Results The NLR and ELR were significantly higher and the LMR was significantly lower
in smokers than nonsmokers. The pack-years were positively correlated with
the NLR and ELR and negatively correlated with the LMR. Conclusion A high NLR and ELR and low LMR are associated with cigarette smoking and may
be useful indicators of systemic inflammation activity, even in healthy
smokers. Smokers with a high NLR and ELR and low LMR can easily be
identified during routine blood analysis and might benefit from preventive
treatment.
Collapse
Affiliation(s)
- Yusuf Çekici
- Department of Cardiology, Gaziantep Dr. Ersin Arslan Education and Research Hospital, Gaziantep, Turkey
| | - Mücahid Yılmaz
- Department of Cardiology, Elazığ Education and Research Hospital, Elazığ, Turkey
| | - Özlem Seçen
- Department of Cardiology, Elazığ Education and Research Hospital, Elazığ, Turkey
| |
Collapse
|
46
|
Andreadou I, Cabrera-Fuentes HA, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, Liehn EA, Gomes CPC, Schulz R, Hausenloy DJ. Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities. Cardiovasc Res 2019; 115:1117-1130. [PMID: 30825305 PMCID: PMC6529904 DOI: 10.1093/cvr/cvz050] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 02/24/2019] [Indexed: 12/22/2022] Open
Abstract
New therapies are required to reduce myocardial infarct (MI) size and prevent the onset of heart failure in patients presenting with acute myocardial infarction (AMI), one of the leading causes of death and disability globally. In this regard, the immune cell response to AMI, which comprises an initial pro-inflammatory reaction followed by an anti-inflammatory phase, contributes to final MI size and post-AMI remodelling [changes in left ventricular (LV) size and function]. The transition between these two phases is critical in this regard, with a persistent and severe pro-inflammatory reaction leading to adverse LV remodelling and increased propensity for developing heart failure. In this review article, we provide an overview of the immune cells involved in orchestrating the complex and dynamic inflammatory response to AMI-these include neutrophils, monocytes/macrophages, and emerging players such as dendritic cells, lymphocytes, pericardial lymphoid cells, endothelial cells, and cardiac fibroblasts. We discuss potential reasons for past failures of anti-inflammatory cardioprotective therapies, and highlight new treatment targets for modulating the immune cell response to AMI, as a potential therapeutic strategy to improve clinical outcomes in AMI patients. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Hector A Cabrera-Fuentes
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Institute of Biochemistry, Medical School, Justus-Liebig University, Ludwigstrasse 23, Giessen, Germany
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Kremlyovskaya St, 18, Kazan, Respublika Tatarstan, Russia
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute Department of Medicine (Cardiology) Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx NY USA
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg, Germany
| | - Tomasz Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Świętej Anny 12, Kraków, Poland
- Institute of Cardiovascular and Medical Sciences, University ofGlasgow, University Avenue, Glasgow, UK
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University,Templergraben 55, Aachen, Germany
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Strada Petru Rareș 2, Craiova, Romania
- Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital, Rheinisch Westfälische Technische Hochschule,Templergraben 55, Aachen, Germany
| | - Clarissa P C Gomes
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Rainer Schulz
- Physiologisches Institut Fachbereich Medizin der Justus-Liebig-Universität, Aulweg 129, Giessen, Germany
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London, UK
| |
Collapse
|
47
|
Kamińska J, Lisowska A, Koper-Lenkiewicz OM, Mikłasz P, Grubczak K, Moniuszko M, Kiszło P, Kemona H, Dymicka-Piekarska V. Differences in Monocyte Subsets and Monocyte-Platelet Aggregates in Acute Myocardial Infarction-PreliminaryResults. Am J Med Sci 2019; 357:421-434. [PMID: 31010468 DOI: 10.1016/j.amjms.2019.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Monocyte-platelet interaction may favor the development of a proatherogenic monocyte phenotype. It is still uncertain which of the 3 monocyte subpopulations interact with platelets to form monocyte-platelet aggregates (MPAs) in acute myocardial infarctions. The aim of our study was to evaluate the monocyte subsets, the percentage of MPAs and the involvement of monocyte subsets in MPA formation among patients with ST-elevation myocardial infarction (STEMI) and non-ST-elevation myocardial infarction (NSTEMI), and compared to patients with stable angina (SA). METHODS Monocyte subsets and MPAs formation were measured in blood collected in 3.2% sodium citrate tubes by means of flow cytometry. RESULTS Classical, intermediate and nonclassical monocyte percentages were statistically different when comparing patients with STEMI and NSTEMI. Moreover, classical and intermediate monocytes were statistically different when comparing the STEMI and SA group; however, only the classical monocyte subset was found to be higher in the acute myocardial infarction group compared to the SA group. The percentage of MPAs was significantly higher in STEMI (50.1%) compared to NSTEMI (22.9%). We found no differences in the involvement of monocyte subsets in MPA formation between patients with STEMI and NSTEMI and in comparison with the SA group. CONCLUSIONS These findings suggest that the increase in circulating levels of classical monocytes in patients with STEMI as compared to NSTEMI reflects the severity of the acute event. The increased percentage of MPAs may favor the development of STEMI compared to NSTEMI.
Collapse
Affiliation(s)
- Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland.
| | - Anna Lisowska
- Department of Cardiology, Clinical Hospital of the Medical University of Bialystok, Bialystok, Poland
| | - Olga M Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Paula Mikłasz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Paweł Kiszło
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Halina Kemona
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | |
Collapse
|
48
|
Kossmann H, Rischpler C, Hanus F, Nekolla SG, Kunze KP, Götze K, Goedel A, Sager H, Kastrati A, Sinnecker D, Kupatt C, Ibrahim T, Schwaiger M, Laugwitz KL, Dirschinger RJ. Monocyte-platelet aggregates affect local inflammation in patients with acute myocardial infarction. Int J Cardiol 2019; 287:7-12. [PMID: 31003796 DOI: 10.1016/j.ijcard.2019.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/01/2019] [Indexed: 01/12/2023]
Abstract
The local inflammatory response following acute myocardial infarction (AMI) is increasingly being recognized as a central factor determining infarct healing. Myocardial inflammation can be visualized in patients using fasting 18F-FDG PET/MRI. Although this novel biosignal correlates with long-term functional outcome, the corresponding cellular substrate is not well understood. Here we present a retrospective analysis of 29 patients with AMI who underwent revascularization, suggesting a connection between post infarction myocardial fasting 18F-FDG uptake, monocyte platelet aggregates (MPA), and P2Y12 inhibition. In detail, patients with high MPA percentages of CD14highCD16+ and CD14lowCD16+ monocytes had significantly higher local 18F-FDG uptake (SUVmean) in the infarcted myocardium than patients with low MPA (p < 0.05). Furthermore, there was an association of high MPA percentage in all monocyte subpopulations with deteriorating ΔLV-EF after 6 months (p < 0.01), which was confirmed in an extended analysis with additional 29 patients without PET/MRI data available. In this analysis, administration of Ticagrelor was associated with lower MPA percentage of CD14high monocyte subpopulations than Clopidogrel (p < 0.01) or Prasugrel (p < 0.05). Taken together, the findings from this analysis suggest that platelet aggregability may affect monocyte extravasation into the infarcted myocardium and influence long-term functional outcome. P2Y12 inhibition may intervene in this pathophysiologic process. Prospective studies are needed to further examine this important relationship.
Collapse
Affiliation(s)
- Hans Kossmann
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christoph Rischpler
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Franziska Hanus
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephan G Nekolla
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Karl P Kunze
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katharina Götze
- Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Goedel
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hendrik Sager
- Deutsches Herzzentrum, Technische Universität München, Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum, Technische Universität München, Munich, Germany
| | - Daniel Sinnecker
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Tareq Ibrahim
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ralf J Dirschinger
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| |
Collapse
|
49
|
Weil BR, Neelamegham S. Selectins and Immune Cells in Acute Myocardial Infarction and Post-infarction Ventricular Remodeling: Pathophysiology and Novel Treatments. Front Immunol 2019; 10:300. [PMID: 30873166 PMCID: PMC6400985 DOI: 10.3389/fimmu.2019.00300] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
The glycosciences aim to understand the impact of extracellular and intracellular carbohydrate structures on biological function. These glycans primarily fall into three major groups: lipid-linked carbohydrates that are referred to as glycosphingolipids or simply glycolipids; relatively short carbohydrate chains that are often O- or N-linked to proteins yielding common glycoproteins; and extended linear polymeric carbohydrate structures that are referred to as glycosaminoglycans (GAGs). Whereas, the impact of such carbohydrate structures has been extensively examined in cancer biology, their role in acute and chronic heart disease is less studied. In this context, a growing body of evidence indicates that glycans play an important role in immune mediated cell recruitment to damaged heart tissue to initiate wound healing and repair after injury. This is particularly important following ischemia and reperfusion that occurs in the heart in the setting of acute myocardial infarction. Here, immune system-mediated repair of the damaged myocardium plays a critical role in determining post-infarction ventricular remodeling, cardiac function, and patient outcome. Further, alterations in immune cell activity can promote the development of heart failure. The present review summarizes our current understanding of the phases of immune-mediated repair following myocardial infarction. It discusses what is known regarding glycans in mediating the recruitment of circulating immune cells during the early inflammatory stage of post-infarction repair, with focus on the selectin family of adhesion molecules. It offers future directions for research aimed at utilizing our knowledge of mechanisms underlying immune cell recruitment to either modulate leukocyte recruitment to the injured tissue or enhance the targeted delivery of biologic therapeutics such as stem cells in an attempt to promote repair of the damaged heart.
Collapse
Affiliation(s)
- Brian R Weil
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Sriram Neelamegham
- Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY, United States.,Department of Chemical & Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, United States
| |
Collapse
|
50
|
Hoedemaker NP, Roolvink V, de Winter RJ, van Royen N, Fuster V, García-Ruiz JM, Er F, Gassanov N, Hanada K, Okumura K, Ibáñez B, van 't Hof AW, Damman P. Early intravenous beta-blockers in patients undergoing primary percutaneous coronary intervention for ST-segment elevation myocardial infarction: A patient-pooled meta-analysis of randomized clinical trials. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2019; 9:469-477. [PMID: 30759994 PMCID: PMC7672673 DOI: 10.1177/2048872619830609] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Conflicting evidence is available on the efficacy and safety of early intravenous beta-blockers before primary percutaneous coronary intervention for ST-segment elevation myocardial infarction. We performed a patient-pooled meta-analysis of trials comparing early intravenous beta-blockers with placebo or routine care in ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention. AIM The aim of this study was to evaluate the clinical and safety outcomes of intravenous beta-blockers in ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention. METHODS Four randomized trials with a total of 1150 patients were included. The main outcome was one-year death or myocardial infarction. Secondary outcomes included biomarker-based infarct size, left ventricular ejection fraction during follow-up, ventricular tachycardia, and a composite safety outcome (cardiogenic shock, symptomatic bradycardia, or hypotension) during hospitalization. RESULTS One-year death or myocardial infarction was similar among beta-blocker (4.2%) and control patients (4.4%) (hazard ratio: 0.96 (95% confidence interval: 0.53-1.75, p=0.90, I2=0%). No difference was observed in biomarker-based infarct size. One-month left ventricular ejection fraction was similar, but left ventricular ejection fraction at six months was significantly higher in patients treated with early intravenous beta-blockade (52.8% versus 50.0% in the control group, p=0.03). No difference was observed in the composite safety outcome or ventricular tachycardia during hospitalization. CONCLUSION In ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention, the administration of early intravenous beta-blockers was safe. However, there was no difference in the main outcome of one-year death or myocardial infarction with early intravenous beta-blockers. A larger clinical trial is warranted to confirm the definitive efficacy of early intravenous beta-blockers.
Collapse
Affiliation(s)
| | | | | | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, The Netherlands
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, USA.,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain
| | - José M García-Ruiz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Fikret Er
- Department of Cardiology and Electrophysiology, Klinikum Gütersloh, Germany
| | - Natig Gassanov
- Department of Cardiology and Electrophysiology, Klinikum Gütersloh, Germany
| | - Kenji Hanada
- Department of Cardiology, Hirosaki University Graduate School of Medicine, Japan
| | - Ken Okumura
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center, Japan
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Spain.,Cardiology Department, IIS-Fundacion Jiménez Díaz University Hospital, Spain
| | - Arnoud W van 't Hof
- Department of Cardiology, Maastricht University Medical Center, The Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, The Netherlands
| |
Collapse
|