51
|
Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev 2021; 198:111540. [PMID: 34237321 PMCID: PMC8387860 DOI: 10.1016/j.mad.2021.111540] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.
Collapse
|
52
|
Katz DH, Tahir UA, Ngo D, Benson MD, Gao Y, Shi X, Nayor M, Keyes MJ, Larson MG, Hall ME, Correa A, Sinha S, Shen D, Herzig M, Yang Q, Robbins JM, Chen ZZ, Cruz DE, Peterson B, Vasan RS, Wang TJ, Wilson JG, Gerszten RE. Multiomic Profiling in Black and White Populations Reveals Novel Candidate Pathways in Left Ventricular Hypertrophy and Incident Heart Failure Specific to Black Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003191. [PMID: 34019435 PMCID: PMC8497179 DOI: 10.1161/circgen.120.003191] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Increased left ventricular (LV) mass is associated with adverse cardiovascular events including heart failure (HF). Both increased LV mass and HF disproportionately affect Black individuals. To understand the underlying mechanisms, we undertook a proteomic screen in a Black cohort and compared the findings to results from a White cohort. METHODS We measured 1305 plasma proteins using the SomaScan platform in 1772 Black participants (mean age, 56 years; 62% women) in JHS (Jackson Heart Study) with LV mass assessed by 2-dimensional echocardiography. Incident HF was assessed in 1600 participants. We then compared protein associations in JHS to those observed in White participants from FHS (Framingham Heart Study; mean age, 54 years; 56% women). RESULTS In JHS, there were 110 proteins associated with LV mass and 13 proteins associated with incident HF hospitalization with false discovery rate <5% after multivariable adjustment. Several proteins showed expected associations with both LV mass and HF, including NT-proBNP (N-terminal pro-B-type natriuretic peptide; β=0.04; P=2×10-8; hazard ratio, 1.48; P=0.0001). The strongest association with LV mass was novel: LKHA4 (leukotriene-A4 hydrolase; β=0.05; P=5×10-15). This association was confirmed on an alternate proteomics platform and further supported by related metabolomic data. Fractalkine/CX3CL1 (C-X3-C Motif Chemokine Ligand 1) showed a novel association with incident HF (hazard ratio, 1.32; P=0.0002). While established biomarkers such as cystatin C and NT-proBNP showed consistent associations in Black and White individuals, LKHA4 and fractalkine were significantly different between the two groups. CONCLUSIONS We identified several novel biological pathways specific to Black adults hypothesized to contribute to the pathophysiologic cascade of LV hypertrophy and incident HF including LKHA4 and fractalkine.
Collapse
Affiliation(s)
- Daniel H. Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Usman A. Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Debby Ngo
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Mark D. Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Yan Gao
- Univ of Mississippi Medical Center, Jackson, MS
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital
| | - Michelle J. Keyes
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Framingham Heart Study, Framingham
| | | | | | | | - Sumita Sinha
- Whitehead Institute for Biomedical Research, Cambridge
| | - Dongxiao Shen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Matthew Herzig
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jeremy M. Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Daniel E. Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Bennet Peterson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Thomas J. Wang
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
53
|
High Neutrophil-to-Lymphocyte Ratio and Platelet-to-Lymphocyte Ratio Are Associated with Poor Survival in Patients with Hemodialysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9958081. [PMID: 34104653 PMCID: PMC8159629 DOI: 10.1155/2021/9958081] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
Background The neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are markers for systemic inflammation condition. Although NLR has emerged as a risk factor for poor survival in end-stage renal disease (ESRD) patients, the relationship between PLR and mortality is still unknown. We aimed to explore the interaction of NLR and PLR in predicting mortality in hemodialysis (HD) patients. Method We enrolled 360 HD patients for a 71-month follow-up. The endpoint was all-cause and cardiovascular (CV) mortality. Pearson correlation analysis was conducted to evaluate the relationship between factors and NLR or PLR. Kaplan-Meier curves and Cox proportional analysis were used to assess the prognostic value of NLR and PLR. Results NLR was positively correlated with neutrophil and negatively correlated with lymphocyte, hemoglobin, and serum albumin. PLR was positively correlated with neutrophil and platelet and negatively correlated with lymphocyte and hemoglobin. In multivariate Cox regression, a higher NLR level was independently associated with all-cause mortality (OR 2.011, 95% CI 1.082-3.74, p = 0.027), while a higher PLR level might predict CV mortality (OR 2.768, 95% CI 1.147-6.677, p = 0.023) in HD patients. Conclusion NLR and PLR are cheap and reliable biomarkers for all-cause and CV mortality to predict survival in HD patients.
Collapse
|
54
|
Pharmacological inhibition of GLUT1 as a new immunotherapeutic approach after myocardial infarction. Biochem Pharmacol 2021; 190:114597. [PMID: 33965393 DOI: 10.1016/j.bcp.2021.114597] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 11/21/2022]
Abstract
Myocardial infarction (MI) is one of the major contributors to cardiovascular morbidity and mortality. Excess inflammation significantly contributes to cardiac remodeling and heart failure after MI. Accumulating evidence has shown the central role of cellular metabolism in regulating the differentiation and function of cells. Metabolic rewiring is particularly relevant for proinflammatory responses induced by ischemia. Hypoxia reduces mitochondrial oxidative phosphorylation (OXPHOS) and induces increased reliance on glycolysis. Moreover, activation of a proinflammatory transcriptional program is associated with preferential glucose metabolism in leukocytes. An improved understanding of the mechanisms that regulate metabolic adaptations holds the potential to identify new metabolic targets and strategies to reduce ischemic cardiac damage, attenuate excess local inflammation and ultimately prevent the development of heart failure. Among possible drug targets, glucose transporter 1 (GLUT1) gained considerable interest considering its pivotal role in regulating glucose availability in activated leukocytes and the availability of small molecules that selectively inhibit it. Therefore, we summarize current evidence on the role of GLUT1 in leukocytes (focusing on macrophages and T cells) and non-leukocytes, including cardiomyocytes, endothelial cells and fibroblasts regarding ischemic heart disease. Beyond myocardial infarction, we can foresee the role of GLUT1 blockers as a possible pharmacological approach to limit pathogenic inflammation in other conditions driven by excess sterile inflammation.
Collapse
|
55
|
Spray L, Park C, Cormack S, Mohammed A, Panahi P, Boag S, Bennaceur K, Sopova K, Richardson G, Stangl VM, Rech L, Rainer PP, Ramos GC, Hofmann U, Stellos K, Spyridopoulos I. The Fractalkine Receptor CX 3CR1 Links Lymphocyte Kinetics in CMV-Seropositive Patients and Acute Myocardial Infarction With Adverse Left Ventricular Remodeling. Front Immunol 2021; 12:605857. [PMID: 34046028 PMCID: PMC8147691 DOI: 10.3389/fimmu.2021.605857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 04/20/2021] [Indexed: 01/10/2023] Open
Abstract
Aims Latent cytomegalovirus (CMV) infection is associated with adverse cardiovascular outcomes. Virus-specific CX3CR1+ effector memory T-cells may be instrumental in this process due to their pro-inflammatory properties. We investigated the role of CX3CR1 (fractalkine receptor) in CMV-related lymphocyte kinetics and cardiac remodeling in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (pPCI). Methods and Results We retrospectively analysed lymphocyte count, troponin, and survival in 4874 STEMI/pPCI patients, evaluated lymphocyte kinetics during reperfusion in a prospective cohort, and obtained sequential cardiac MRI (cMRI) to assess remodeling. Pre-reperfusion lymphopenia independently predicted mortality at 7.5 years. Prior to reperfusion, CCR7+ T-lymphocytes appeared to be depleted. After reperfusion, T-lymphocytes expressing CX3CR1 were depleted predominantly in CMV-seropositive patients. During ischaemia/reperfusion, a drop in CX3CR1+ T-lymphocytes was significantly linked with microvascular obstruction in CMV+ patients, suggesting increased fractalkine-receptor interaction. At 12 weeks, CMV+ patients displayed adverse LV remodeling. Conclusion We show that lymphopenia occurs before and after reperfusion in STEMI by different mechanisms and predicts long-term outcome. In CMV+ patients, increased fractalkine induction and sequestration of CX3CR1+ T-cells may contribute to adverse remodeling, suggesting a pro-inflammatory pathomechanism which presents a novel therapeutic target.
Collapse
Affiliation(s)
- Luke Spray
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Catherine Park
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Suzanne Cormack
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Ashfaq Mohammed
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Pedram Panahi
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Stephen Boag
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Karim Bennaceur
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Kateryna Sopova
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gavin Richardson
- Biosciences Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Verena M. Stangl
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Lavinia Rech
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Gustavo Campos Ramos
- Department of Internal Medicine I and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Ulrich Hofmann
- Department of Internal Medicine I and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Konstantinos Stellos
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
- Biosciences Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Ioakim Spyridopoulos
- Cardiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Cardiovascular Biology and Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
56
|
Marsh SA, Park C, Redgrave RE, Singh E, Draganova L, Boag SE, Spray L, Ali S, Spyridopoulos I, Arthur HM. Rapid fall in circulating non-classical monocytes in ST elevation myocardial infarction patients correlates with cardiac injury. FASEB J 2021; 35:e21604. [PMID: 33913566 DOI: 10.1096/fj.202100240r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 11/11/2022]
Abstract
Myocardial infarction leads to a rapid innate immune response that is ultimately required for repair of damaged heart tissue. We therefore examined circulating monocyte dynamics immediately after reperfusion of the culprit coronary vessel in STEMI patients to determine whether this correlated with level of cardiac injury. A mouse model of cardiac ischemia/reperfusion injury was subsequently used to establish the degree of monocyte margination to the coronary vasculature that could potentially contribute to the drop in circulating monocytes. We retrospectively analyzed blood samples from 51 STEMI patients to assess the number of non-classical (NC), classical, and intermediate monocytes immediately following primary percutaneous coronary intervention. Classical and intermediate monocytes showed minimal change. On the other hand, circulating numbers of NC monocytes fell by approximately 50% at 90 minutes post-reperfusion. This rapid decrease in NC monocytes was greatest in patients with the largest infarct size (P < .05) and correlated inversely with left ventricular function (r = 0.41, P = .04). The early fall in NC monocytes post-reperfusion was confirmed in a second prospective study of 13 STEMI patients. Furthermore, in a mouse cardiac ischemia model, there was significant monocyte adhesion to coronary vessel endothelium at 2 hours post-reperfusion pointing to a specific and rapid vessel margination response to cardiac injury. In conclusion, rapid depletion of NC monocytes from the circulation in STEMI patients following coronary artery reperfusion correlates with the level of acute cardiac injury and involves rapid margination to the coronary vasculature.
Collapse
Affiliation(s)
- Sarah A Marsh
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Catherine Park
- Translational and Clinical Research Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Rachael E Redgrave
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Esha Singh
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Lilia Draganova
- Translational and Clinical Research Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Stephen E Boag
- Translational and Clinical Research Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Luke Spray
- Cardiology Department, Freeman Hospital, Newcastle, UK
| | - Simi Ali
- Translational and Clinical Research Institute, Leech Building, Newcastle University, Newcastle, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Centre for Life, Newcastle University, Newcastle, UK
| | - Helen M Arthur
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle, UK
| |
Collapse
|
57
|
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
|
58
|
Kologrivova I, Shtatolkina M, Suslova T, Ryabov V. Cells of the Immune System in Cardiac Remodeling: Main Players in Resolution of Inflammation and Repair After Myocardial Infarction. Front Immunol 2021; 12:664457. [PMID: 33868315 PMCID: PMC8050340 DOI: 10.3389/fimmu.2021.664457] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
The burden of heart failure (HF), developing after myocardial infarction MI, still represents a major issue in clinical practice. Failure of appropriate resolution of inflammation during post-myocardial injury is associated with unsuccessful left ventricular remodeling and underlies HF pathogenesis. Cells of the immune system have been shown to mediate both protective and damaging effects in heart remodeling. This ambiguity of the role of the immune system and inconsistent results of the recent clinical trials question the benefits of anti-inflammatory therapies during acute MI. The present review will summarize knowledge of the roles that different cells of the immune system play in the process of post-infarct cardiac healing. Data on the phenotype, active molecules and functions of the immune cells, based on the results of both experimental and clinical studies, will be provided. For some cellular subsets, such as macrophages, neutrophils, dendritic cells and lymphocytes, an anti-inflammatory activity has been attributed to the specific subpopulations. Activity of other cells, such as eosinophils, mast cells, natural killer (NK) cells and NKT cells has been shown to be highly dependent of the signals created by micro-environment. Also, new approaches for classification of cellular phenotypes based on the single-cell RNA sequencing allow better understanding of the phenotype of the cells involved in resolution of inflammation. Possible perspectives of immune-mediated therapy for AMI patients are discussed in the conclusion. We also outline unresolved questions that need to be solved in order to implement the current knowledge on the role of the immune cells in post-MI tissue repair into practice.
Collapse
Affiliation(s)
- Irina Kologrivova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Shtatolkina
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Tatiana Suslova
- Department of Clinical Laboratory Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Vyacheslav Ryabov
- Department of Emergency Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia.,Division of Cardiology, Department of Professional Development and Retraining, Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
59
|
Santos-Zas I, Lemarié J, Zlatanova I, Cachanado M, Seghezzi JC, Benamer H, Goube P, Vandestienne M, Cohen R, Ezzo M, Duval V, Zhang Y, Su JB, Bizé A, Sambin L, Bonnin P, Branchereau M, Heymes C, Tanchot C, Vilar J, Delacroix C, Hulot JS, Cochain C, Bruneval P, Danchin N, Tedgui A, Mallat Z, Simon T, Ghaleh B, Silvestre JS, Ait-Oufella H. Cytotoxic CD8 + T cells promote granzyme B-dependent adverse post-ischemic cardiac remodeling. Nat Commun 2021; 12:1483. [PMID: 33674611 PMCID: PMC7935973 DOI: 10.1038/s41467-021-21737-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022] Open
Abstract
Acute myocardial infarction is a common condition responsible for heart failure and sudden death. Here, we show that following acute myocardial infarction in mice, CD8+ T lymphocytes are recruited and activated in the ischemic heart tissue and release Granzyme B, leading to cardiomyocyte apoptosis, adverse ventricular remodeling and deterioration of myocardial function. Depletion of CD8+ T lymphocytes decreases apoptosis within the ischemic myocardium, hampers inflammatory response, limits myocardial injury and improves heart function. These effects are recapitulated in mice with Granzyme B-deficient CD8+ T cells. The protective effect of CD8 depletion on heart function is confirmed by using a model of ischemia/reperfusion in pigs. Finally, we reveal that elevated circulating levels of GRANZYME B in patients with acute myocardial infarction predict increased risk of death at 1-year follow-up. Our work unravels a deleterious role of CD8+ T lymphocytes following acute ischemia, and suggests potential therapeutic strategies targeting pathogenic CD8+ T lymphocytes in the setting of acute myocardial infarction. Immune cells contribute to adverse remodeling following myocardial infarction. Here the authors show in mice and pigs that CD8+ lymphocytes release Granzyme B in the infarcted heart leading to cardiomyocyte death, enhanced inflammation and deterioration of cardiac function.
Collapse
Affiliation(s)
| | | | | | - Marine Cachanado
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | | | - Hakim Benamer
- Service de cardiologie, Institut Cardiovasculaire Paris Sud, Paris, France
| | - Pascal Goube
- Service de cardiologie, Centre Hospitalier de Corbeil, Corbeil, France
| | | | - Raphael Cohen
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Maya Ezzo
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Vincent Duval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Yujiao Zhang
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Jin-Bo Su
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Alain Bizé
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Lucien Sambin
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Philippe Bonnin
- Inserm U965, Department of Physiology, Assistance Publique Hôpitaux de Paris, Hôpital Lariboisière, France
| | - Maxime Branchereau
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Christophe Heymes
- Inserm U1048-Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | | | - José Vilar
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | | | | | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Bruneval
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Service d'anatomopathologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Nicolas Danchin
- Service de cardiologie, Hôpital Europeen G. Pompidou, Assistance Publique, Hôpitaux de Paris, Paris, France
| | - Alain Tedgui
- Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - Ziad Mallat
- Université de Paris, PARCC, INSERM, F-75015, Paris, France.,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | - Tabassome Simon
- Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France.,Sorbonne Université, UPMC-site St Antoine, Service de Pharmacologie, Assistance Publique-Hôpitaux de Paris, APHP.SU; Department of Clinical Pharmacology and Clinical Research Platform (URCEST-CRB-CRC-EST), Hôpital Saint Antoine, Paris, France
| | - Bijan Ghaleh
- Inserm U955-IMRB, Equipe 03, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Hafid Ait-Oufella
- Université de Paris, PARCC, INSERM, F-75015, Paris, France. .,Sorbonne Université, Service de médecine intensive-Réanimation, Assistance Publique, Hôpitaux de Paris, Paris, France.
| |
Collapse
|
60
|
de Dios E, Rios-Navarro C, Pérez-Solé N, Gavara J, Marcos-Garcés V, Forteza MJ, Oltra R, Vila JM, Chorro FJ, Bodi V. Overexpression of genes involved in lymphocyte activation and regulation are associated with reduced CRM-derived cardiac remodelling after STEMI. Int Immunopharmacol 2021; 95:107490. [PMID: 33677257 DOI: 10.1016/j.intimp.2021.107490] [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: 12/16/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 11/19/2022]
Abstract
AIMS Lymphopenia after ST-segment elevation myocardial infarction (STEMI) correlates with deleterious cardiac consequences and worse prognosis. An in-depth examination of genes implicated in lymphocyte proliferation, activation and regulation and their association with short- and long-term cardiac structure and function is therefore of great interest. METHODS Peripheral blood mononuclear cells were isolated from 10 control subjects and 64 patients with a first STEMI treated with primary percutaneous coronary intervention and submitted to cardiac magnetic resonance after 1 week and 6 months. mRNA expression of genes implicated in lymphocyte activation (CD25 and CD69) and regulation [programmed death (PD)-1 and cytotoxic T-lymphocyte antigen (CTLA)-4] were determined by qRT-PCR. RESULTS In comparison to controls, STEMI patients showed heightened mRNA expression of CD25 and lower PD-1 and CTLA-4 96 h after coronary reperfusion. Patients with extensive infarctions (>30% of left ventricular mass) at 1 week displayed a notable reduction in CD25, CD69, PD-1, and CTLA-4 expression (p < 0.05). However, CD25 was the only predictor of 1-week extensive infarct size in multivariate logistic regression analysis (odds ratio 0.019; 95% confidence interval [0.001-0.505]; p = 0.018). Regarding long-term ventricular function, mRNA expression of CD25 under the mean value was associated with worse ventricular function and more adverse remodelling. CONCLUSIONS Following STEMI, heightened expression of genes expressed in regulatory T cells (CD25 and CD69) and immune checkpoints (PD-1 and CTLA-4) correlates with a better short- and long-term cardiac structure and function. Advancing understanding of the pathophysiology of lymphopenia and evaluating novel immunomodulatory therapies will help translate these results into future clinical trials.
Collapse
Affiliation(s)
- Elena de Dios
- Centro de Investigación Biomédica en Red - Cardiovascular (CIBER-CV), 28029 Madrid, Spain; Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | | | | | - Jose Gavara
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain
| | | | - Maria J Forteza
- Cardiovascular Medicine Unit, Center of Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Ricardo Oltra
- Intensive Care Unit, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - José M Vila
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; Physiology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Francisco J Chorro
- Centro de Investigación Biomédica en Red - Cardiovascular (CIBER-CV), 28029 Madrid, Spain; Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Institute of Health Research-INCLIVA, 46010 Valencia, Spain; Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Vicente Bodi
- Centro de Investigación Biomédica en Red - Cardiovascular (CIBER-CV), 28029 Madrid, Spain; Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Institute of Health Research-INCLIVA, 46010 Valencia, Spain; Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain.
| |
Collapse
|
61
|
Zang X, Zhou J, Zhang X, Han Y, Chen X. Ischemia Reperfusion Injury: Opportunities for Nanoparticles. ACS Biomater Sci Eng 2020; 6:6528-6539. [PMID: 33320610 DOI: 10.1021/acsbiomaterials.0c01197] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ischemia reperfusion (IR)-induced oxidative stress, accompanied by inflammatory responses, contributes to morbidity and mortality in numerous diseases such as acute coronary syndrome, stroke, organ transplantation, and limb injury. Ischemia results in profound hypoxia and tissue dysfunction, whereas subsequent reperfusion further aggravates ischemic tissue damage through inducing cell death and activating inflammatory responses. In this review, we highlight recent studies of therapeutic strategies against IR injury. Furthermore, nanotechnology offers significant improvements in this area. Hence, we also review recent advances in nanomedicines for IR therapy, suggesting them as potent and promising strategies to improve drug delivery to IR-injured tissues and achieve protective effects.
Collapse
Affiliation(s)
- Xinlong Zang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Jingyi Zhou
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Xiaoxu Zhang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Yantao Han
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| |
Collapse
|
62
|
de Dios E, Rios-Navarro C, Perez-Sole N, Gavara J, Marcos-Garces V, Rodríguez E, Carratalá A, Forner MJ, Navarro J, Blasco ML, Bondia E, Signes-Costa J, Vila JM, Forteza MJ, Chorro FJ, Bodi V. Similar Clinical Course and Significance of Circulating Innate and Adaptive Immune Cell Counts in STEMI and COVID-19. J Clin Med 2020; 9:jcm9113484. [PMID: 33126723 PMCID: PMC7692467 DOI: 10.3390/jcm9113484] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
This study aimed to assess the time course of circulating neutrophil and lymphocyte counts and their ratio (NLR) in ST-segment elevation myocardial infarction (STEMI) and coronavirus disease (COVID)-19 and explore their associations with clinical events and structural damage. Circulating neutrophil, lymphocyte and NLR were sequentially measured in 659 patients admitted for STEMI and in 103 COVID-19 patients. The dynamics detected in STEMI (within a few hours) were replicated in COVID-19 (within a few days). In both entities patients with events and with severe structural damage displayed higher neutrophil and lower lymphocyte counts. In both scenarios, higher maximum neutrophil and lower minimum lymphocyte counts were associated with more events and more severe organ damage. NLR was higher in STEMI and COVID-19 patients with the worst clinical and structural outcomes. A canonical deregulation of the immune response occurs in STEMI and COVID-19 patients. Boosted circulating innate (neutrophilia) and depressed circulating adaptive immunity (lymphopenia) is associated with more events and severe organ damage. A greater understanding of these critical illnesses is pivotal to explore novel alternative therapies.
Collapse
Affiliation(s)
- Elena de Dios
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Cesar Rios-Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | - Nerea Perez-Sole
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | - Jose Gavara
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
| | | | - Enrique Rodríguez
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Biochemical Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Arturo Carratalá
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Biochemical Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Maria J. Forner
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Internal Medicine Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Jorge Navarro
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Medical Directory, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Maria L. Blasco
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Medical Intensive Care Unit, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Elvira Bondia
- Pneumology Service, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Jaime Signes-Costa
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Pneumology Service, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Jose M. Vila
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Physiology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Maria J. Forteza
- Cardiovascular Medicine Unit, Center of Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden;
| | - Francisco J. Chorro
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain;
| | - Vicente Bodi
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain; (E.d.D.); (F.J.C.)
- Medicine Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.R.-N.); (N.P.-S.); (J.G.); (E.R.); (A.C.); (J.N.); (M.L.B.); (J.S.-C.); (J.M.V.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain;
- Correspondence: ; Tel.: +34-96-197-3523
| |
Collapse
|
63
|
Abstract
Several members of the chemokine family are involved in regulation of fibrosis. This review manuscript discusses the role of the chemokines in the pathogenesis of myocardial fibrosis. The CC chemokine CCL2 exerts fibrogenic actions through recruitment and activation of monocytes and macrophages expressing its receptor, CCR2. Other CC chemokines may also contribute to fibrotic remodeling by recruiting subsets of fibrogenic macrophages. CXC chemokines containing the ELR motif may exert pro-fibrotic actions, through recruitment of activated neutrophils and subsequent formation of neutrophil extracellular traps (NETs), or via activation of fibrogenic monocytes. CXCL12 has also been suggested to exert fibrogenic actions through effects on fibroblasts and immune cells. In contrast, the CXCR3 ligand CXCL10 was found to reduce cardiac fibrosis, inhibiting fibroblast migration. Chemokines are critical links between inflammation and fibrosis in myocardial disease and may be promising therapeutic targets for patients with heart failure accompanied by prominent inflammation and fibrosis.
Collapse
Affiliation(s)
- Ruoshui Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
64
|
Dookun E, Walaszczyk A, Redgrave R, Palmowski P, Tual‐Chalot S, Suwana A, Chapman J, Jirkovsky E, Donastorg Sosa L, Gill E, Yausep OE, Santin Y, Mialet‐Perez J, Andrew Owens W, Grieve D, Spyridopoulos I, Taggart M, Arthur HM, Passos JF, Richardson GD. Clearance of senescent cells during cardiac ischemia-reperfusion injury improves recovery. Aging Cell 2020; 19:e13249. [PMID: 32996233 PMCID: PMC7576252 DOI: 10.1111/acel.13249] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 12/16/2022] Open
Abstract
A key component of cardiac ischemia-reperfusion injury (IRI) is the increased generation of reactive oxygen species, leading to enhanced inflammation and tissue dysfunction in patients following intervention for myocardial infarction. In this study, we hypothesized that oxidative stress, due to ischemia-reperfusion, induces senescence which contributes to the pathophysiology of cardiac IRI. We demonstrate that IRI induces cellular senescence in both cardiomyocytes and interstitial cell populations and treatment with the senolytic drug navitoclax after ischemia-reperfusion improves left ventricular function, increases myocardial vascularization, and decreases scar size. SWATH-MS-based proteomics revealed that biological processes associated with fibrosis and inflammation that were increased following ischemia-reperfusion were attenuated upon senescent cell clearance. Furthermore, navitoclax treatment reduced the expression of pro-inflammatory, profibrotic, and anti-angiogenic cytokines, including interferon gamma-induced protein-10, TGF-β3, interleukin-11, interleukin-16, and fractalkine. Our study provides proof-of-concept evidence that cellular senescence contributes to impaired heart function and adverse remodeling following cardiac ischemia-reperfusion. We also establish that post-IRI the SASP plays a considerable role in the inflammatory response. Subsequently, senolytic treatment, at a clinically feasible time-point, attenuates multiple components of this response and improves clinically important parameters. Thus, cellular senescence represents a potential novel therapeutic avenue to improve patient outcomes following cardiac ischemia-reperfusion.
Collapse
Affiliation(s)
- Emily Dookun
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Anna Walaszczyk
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | - Pawel Palmowski
- School of Environmental SciencesFaculty of ScienceAgriculture & EngineeringNewcastle UniversityNewcastle upon TyneUK
| | | | - Averina Suwana
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - James Chapman
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | | | - Eleanor Gill
- School of MedicineDentistry and Biomedical SciencesCentre for Experimental MedicineInstitute for Health SciencesQueen`s University BelfastBelfastUK
| | - Oliver E Yausep
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | | | - W Andrew Owens
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - David Grieve
- School of MedicineDentistry and Biomedical SciencesCentre for Experimental MedicineInstitute for Health SciencesQueen`s University BelfastBelfastUK
| | | | - Michael Taggart
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Helen M. Arthur
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - João F. Passos
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| | | |
Collapse
|
65
|
Vasilieva E, Gianella S, Freeman ML. Novel Strategies to Combat CMV-Related Cardiovascular Disease. Pathog Immun 2020; 5:240-274. [PMID: 33089035 PMCID: PMC7556413 DOI: 10.20411/pai.v5i1.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV), a ubiquitous human pathogen that is never cleared from the host, has long been thought to be relatively innocuous in immunocompetent adults, but causes severe complications including blindness, end-organ disease, and death in newborns and in immuno-compromised individuals, such as organ transplant recipients and those suffering from AIDS. Yet even in persons with intact immunity, CMV infection is associated with profound stimulation of immune and inflammatory pathways. Carriers of CMV infection also have an elevated risk of developing cardiovascular complications. In this review, we define the proposed mechanisms of how CMV contributes to cardiovascular disease (CVD), describe current approaches to target CMV, and discuss how these strategies may or may not alleviate cardiovascular complications in those with CMV infection. In addition, we discuss the special situation of CMV coinfection in people with HIV infection receiving antiretroviral therapy, and describe how these 2 viral infections may interact to potentiate CVD in this especially vulnerable population.
Collapse
Affiliation(s)
- Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine; Department of Medicine; Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
66
|
Bradshaw AD, DeLeon-Pennell KY. T-cell regulation of fibroblasts and cardiac fibrosis. Matrix Biol 2020; 91-92:167-175. [PMID: 32438054 PMCID: PMC7434661 DOI: 10.1016/j.matbio.2020.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.
Collapse
Affiliation(s)
- Amy D Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States.
| |
Collapse
|
67
|
Deng Z, Zhang M, Zhu T, Zhili N, Liu Z, Xiang R, Zhang W, Xu Y. Dynamic changes in peripheral blood lymphocyte subsets in adult patients with COVID-19. Int J Infect Dis 2020; 98:353-358. [PMID: 32634585 PMCID: PMC7334931 DOI: 10.1016/j.ijid.2020.07.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/14/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19), caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread widely. The aim of this study was to investigate the dynamic changes in peripheral blood lymphocyte subsets in adult patients with COVID-19. METHODS The electronic medical records were reviewed. Data including demographic characteristics, clinical manifestations, comorbidities, laboratory data, and radiological examinations of 435 hospitalized COVID-19 patients with a confirmed SARS-CoV-2 viral infection were extracted and analyzed retrospectively. Lymphocyte subset counts at each week after the onset of the illness were compared with those of the other weeks of illness and with those of control individuals. RESULTS The various lymphocyte subsets (CD3+, CD4+, CD8+, CD19+, and CD16/56+) were below the normal ranges at 1 week after the onset of illness, reaching a nadir during the second week. They increased gradually during the third week and returned to normal levels in the fifth week, but were still lower than those of the healthy controls. The CD3+, CD4+, and CD8+ counts were significantly lower in patients with severe disease compared to those with non-severe disease, and in patients who died compared to those who recovered. DISCUSSION This research indicates that the levels of peripheral blood lymphocyte subsets (CD3+, CD4+, and CD8+) are associated with disease progression and severity, and with the prognosis in patients with COVID-19. Dynamic monitoring of human immune function is one of the indicators for evaluating the severity of disease and the prognosis of COVID-19 patients, and is useful for formulating appropriate treatment strategies.
Collapse
Affiliation(s)
- Zhifeng Deng
- Department of Otolaryngology - Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Minli Zhang
- Yaxin School of Nursing, Wuhan Institute of Design and Sciences, Wuhan 430205, China
| | - Ting Zhu
- Department of Otolaryngology - Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Niu Zhili
- Department of Laboratory Science, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zheming Liu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rong Xiang
- Department of Otolaryngology - Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei Zhang
- Department of Otolaryngology - Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yu Xu
- Department of Otolaryngology - Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
68
|
Hu X, Wu T, Wang C, Li J, Ying C. CD248+CD8+ T lymphocytes suppress pathological vascular remodeling in human thoracic aortic aneurysms. Exp Biol Med (Maywood) 2020; 246:121-129. [PMID: 32867546 DOI: 10.1177/1535370220953386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aortic aneurysms are characterized by vascular inflammation, neovascularization, and extracellular matrix destruction of the aortic wall. Although experimental studies indicate a potential role of CD248 in microvessel remodeling, the functions of CD248 in human vascular pathologies remain unexplored. Here we aimed to study how CD248 interferes with pathological vascular remodeling of human aortic aneurysms. Immunofluorescent staining showed that CD248 expression was mainly localized in the CD8+ T cells infiltrating in the adventitia and media of aortic walls of patients with ascending thoracic aortic aneurysms. qPCR and immunofluorescent staining analyses revealed increased aortic CD248 expression and infiltrating CD248+CD8+ T cells in aortic aneurysms than in nonaneurysmal aortas. Flow cytometry analysis of human peripheral blood further identified a fraction of circulating CD248+ cells which was confined in the CD8+ T-cell compartment. The increased infiltrating of CD248+CD8+ T cells was coincident with reduced circulating CD248+CD8+ T cells in patients with ascending TAA when compared with patients with coronary artery diseases and healthy donors. The CD248+CD8+ T cells were characterized by upregulated IL-10 and downregulated IL-1β/INF-γ expression when compared with CD248-CD8+ T cells. Moreover, when co-cultured with human aortic endothelial cells, the CD248+CD8+ T cells not only downregulated endothelial expression of ICAM1/VCAM1 and MMP2/3 but also suppressed endothelial migration. This study shows that CD248 reduces pathological vascular remodeling via anti-inflammatory CD248+CD8+ T cells, revealing a CD248-mediated cellular mechanism against human aortic aneurysms.
Collapse
Affiliation(s)
- Xiaojuan Hu
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Ting Wu
- Renji Clinical Stem Cell Research Center, Shanghai 200127, China
| | - Chenxi Wang
- Department of Cardiovascular Surgery, School of Medicine, Shanghai Jiao Tong University, Ren Ji Hospital, Shanghai 200127, China
| | - Jun Li
- Renji Clinical Stem Cell Research Center, Shanghai 200127, China
| | - Chunmei Ying
- Department of Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| |
Collapse
|
69
|
Yang T, Guo L, Chen L, Li J, Li Q, Pi Y, Zhu J, Zhang L. A novel role of FKN/CX3CR1 in promoting osteogenic transformation of VSMCs and atherosclerotic calcification. Cell Calcium 2020; 91:102265. [PMID: 32814243 DOI: 10.1016/j.ceca.2020.102265] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 11/19/2022]
Abstract
Fractalkine (FKN) and its specific receptor CX3CR1 play a critical role in the pathogenesis of atherosclerosis including recruitment of vascular cells and the development of inflammation. However, its contribution to regulating the development of atherosclerotic calcification has not been well documented. Osteogenic transformation of vascular smooth muscle cells (VSMCs) is critical in the development of calcification in atherosclerotic lesions. In this study, for the first time, we evaluated the effect of FKN/CX3CR1 on the progression of VSMCs calcification and defined molecular signaling that is operative in the FKN/CX3CR1-induced osteogenic transformation of VSMCs. We found that high-fat diet induced atherosclerotic calcification in vivo was markedly inhibited in the Apolipoprotein E (ApoE) and CX3CR1 deficient (ApoE-/-/CX3CR1-/-) mice compared with their control littermates. FKN and CX3CR1 were both expressed in VSMCs and up-regulated by oxidized low-density lipoprotein (ox-LDL). FKN/CX3CR1 promoted the expression of osteogenic markers, including osteopontin (OPN), bone morphogenetic protein (BMP)-2 and alkaline phosphatase (ALP) and decreased VSMCs markers, including smooth muscle (SM) α-actin and SM22-α in a dose-dependent manner. The essential role of FKN/CX3CR1 in VSMCs calcification was further confirmed by lentivirus-mediated knockdown or overexpression of CX3CR1 blocked or accelerated osteogenic transformation of VSMCs. This response was associated with reciprocal up- and down-regulation of osteogenic factor, runt-related transcription factor 2 (RUNX2), transcription factors in osteoclast differentiation, receptor activator of nuclear factor-κB (RANK), RANK ligand (RNAKL) and osteoprotegerin (OPG), respectively. Inhibition of FKN/CX3CR1-activated Jak2/Stat3 signaling by the Jak/Stat inhibitor AG490 blocked osteogenic transformation of VSMCs and RUNX2 induction concurrently. Taken together, our data uncovered novel roles of FKN/CX3CR1 in promoting VSMC osteogenic transformation and atherosclerotic calcification by activating RUNX2 through Jak2/Stat3 signaling pathway and suppressing OPG. Our findings suggest that targeting FKN/CX3CR1 may provide new strategies for the prevention and treatment of atherosclerotic calcification.
Collapse
Affiliation(s)
- Tong Yang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Lu Guo
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Lizhao Chen
- Department of Neurosurgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Jingcheng Li
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Qiong Li
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Yan Pi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Jie Zhu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Lili Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| |
Collapse
|
70
|
Effect of Interleukin-17 in the Activation of Monocyte Subsets in Patients with ST-Segment Elevation Myocardial Infarction. J Immunol Res 2020; 2020:5692829. [PMID: 32676508 PMCID: PMC7336211 DOI: 10.1155/2020/5692829] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/12/2020] [Accepted: 05/21/2020] [Indexed: 11/18/2022] Open
Abstract
Interleukin- (IL-) 17 is increased in acute myocardial infarction (AMI) and plays a key role in inflammatory diseases through its involvement in the activation of leukocytes. Here, we describe for the first time the effect of IL-17 in the migration and activation of monocyte subsets in patients during ST-segment elevation myocardial infarction (STEMI) and post-STEMI. We analyzed the circulating levels of IL-17 in patient plasma. A gradual increase in IL-17 was found in STEMI and post-STEMI patients. Additionally, IL-17 had a powerful effect on the recruitment of CD14++CD16+/CD14+CD16++ monocytes derived from patients post-STEMI compared with the monocytes from patients with STEMI, suggesting that IL-17 recruits monocytes with inflammatory activity post-STEMI. Furthermore, IL-17 increased the expression of TLR4 on CD14 + CD16 - and CD14++CD16+/CD14+CD16++ monocytes post-STEMI and might enhance the response to danger-associated molecular patterns post-STEMI. Moreover, IL-17 induced secretion of IL-6 from CD14++CD16- and CD14++CD16+/CD14+CD16++ monocytes both in STEMI and in post-STEMI, which indicates that IL-17 has an effect on the secretion of proinflammatory cytokines from monocytes during STEMI and post-STEMI. Overall, we demonstrate that in STEMI and post-STEMI, IL-17 is increased and induces the migration and activation of monocyte subsets, possibly contributing to the inflammatory response through TLR4 and IL-6 secretion.
Collapse
|
71
|
Abstract
In the infarcted myocardium, cardiomyocyte necrosis triggers an intense inflammatory reaction that not only is critical for cardiac repair, but also contributes to adverse remodeling and to the pathogenesis of heart failure. Both CC and CXC chemokines are markedly induced in the infarcted heart, bind to endothelial glycosaminoglycans, and regulate leukocyte trafficking and function. ELR+ CXC chemokines (such as CXCL8) control neutrophil infiltration, whereas CC chemokines (such as CCL2) mediate recruitment of mononuclear cells. Moreover, some members of the chemokine family (such as CXCL10 and CXCL12) may mediate leukocyte-independent actions, directly modulating fibroblast and vascular cell function. This review manuscript discusses our understanding of the role of the chemokines in regulation of injury, repair, and remodeling following myocardial infarction. Although several chemokines may be promising therapeutic targets in patients with myocardial infarction, clinical implementation of chemokine-based therapeutics is hampered by the broad effects of the chemokines in both injury and repair.
Collapse
|
72
|
Chen B, Morris SR, Panigrahi S, Michaelson GM, Wyrick JM, Komissarov AA, Potashnikova D, Lebedeva A, Younes SA, Harth K, Kashyap VS, Vasilieva E, Margolis L, Zidar DA, Sieg SF, Shive CL, Funderburg NT, Gianella S, Lederman MM, Freeman ML. Cytomegalovirus Coinfection Is Associated with Increased Vascular-Homing CD57 + CD4 T Cells in HIV Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2722-2733. [PMID: 32229536 PMCID: PMC7315224 DOI: 10.4049/jimmunol.1900734] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.
Collapse
Affiliation(s)
- Bonnie Chen
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Stephen R Morris
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Soumya Panigrahi
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Gillian M Michaelson
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Jonathan M Wyrick
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Alexey A Komissarov
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Daria Potashnikova
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
- Department of Cell Biology and Histology, School of Biology, Moscow State University, Moscow 119234, Russia
| | - Anna Lebedeva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Souheil-Antoine Younes
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Karem Harth
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Vikram S Kashyap
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Leonid Margolis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - David A Zidar
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH 44106
| | - Scott F Sieg
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Carey L Shive
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH 43210; and
| | - Sara Gianella
- Center for AIDS Research, Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Michael M Lederman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Michael L Freeman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH 44106;
| |
Collapse
|
73
|
Mourouzis K, Oikonomou E, Siasos G, Tsalamadris S, Vogiatzi G, Antonopoulos A, Fountoulakis P, Goliopoulou A, Papaioannou S, Tousoulis D. Pro-inflammatory Cytokines in Acute Coronary Syndromes. Curr Pharm Des 2020; 26:4624-4647. [PMID: 32282296 DOI: 10.2174/1381612826666200413082353] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Over the last decades, the role of inflammation and immune system activation in the initiation and progression of coronary artery disease (CAD) has been established. OBJECTIVES The study aimed to present the interplay between cytokines and their actions preceding and shortly after ACS. METHODS We searched in a systemic manner the most relevant articles to the topic of inflammation, cytokines, vulnerable plaque and myocardial infarction in MEDLINE, COCHRANE and EMBASE databases. RESULTS Different classes of cytokines (intereleukin [IL]-1 family, Tumor necrosis factor-alpha (TNF-α) family, chemokines, adipokines, interferons) are implicated in the entire process leading to destabilization of the atherosclerotic plaque, and consequently, to the incidence of myocardial infarction. Especially IL-1 and TNF-α family are involved in inflammatory cell accumulation, vulnerable plaque formation, platelet aggregation, cardiomyocyte apoptosis and adverse remodeling following the myocardial infarction. Several cytokines such as IL-6, adiponectin, interferon-γ, appear with significant prognostic value in ACS patients. Thus, research interest focuses on the modulation of inflammation in ACS to improve clinical outcomes. CONCLUSION Understanding the unique characteristics that accompany each cytokine-cytokine receptor interaction could illuminate the signaling pathways involved in plaque destabilization and indicate future treatment strategies to improve cardiovascular prognosis in ACS patients.
Collapse
Affiliation(s)
- Konstantinos Mourouzis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sotiris Tsalamadris
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgia Vogiatzi
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alexios Antonopoulos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Petros Fountoulakis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Athina Goliopoulou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Spyridon Papaioannou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
74
|
Luo R, Guo SM, Li YQ, Yang Y, Li ML, Han M, He XF, Ge SW, Xu G. Plasma fractalkine levels are associated with renal inflammation and outcomes in immunoglobulin A nephropathy. Nephrol Dial Transplant 2020; 34:1549-1558. [PMID: 30010903 DOI: 10.1093/ndt/gfy169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND A recognized noninvasive biomarker to improve risk stratification of immunoglobulin A nephropathy (IgAN) patients is scarce. Fractalkine has been shown to play a key role in glomerular disease as chemoattractant, adhesion and even fibrosis factor. The current study assessed the possibility of plasma fractalkine as a novel biomarker in IgAN patients. METHODS Plasma fractalkine was measured in 229 patients with renal biopsy consistent IgAN from 2012 to 2014, and clinical, pathological and prognostic relationships were analyzed. RESULTS The plasma fractalkine levels in IgAN patients were significantly correlated with the creatinine level and 24-h urine protein by both univariate and multivariate analysis. Mesangial hypercellularity was still significantly correlated with the plasma fractalkine levels even after adjustment for other potential predictor variables by multivariate analysis. In addition, the counts of CD20+ B cells or CD68+ macrophage in renal biopsies of IgAN patients were significantly correlated with the plasma fractalkine levels, but not CD4+ and CD8+ T cells. Finally, we concluded that patients with higher plasma fractalkine levels had higher risk of poor renal outcome compared with those with lower plasma fractalkine levels. No association was observed between the CX3CR1 polymorphisms and clinical parameters including plasma fractalkine levels and prognosis. Recombinant fractalkine induced mesangial cells extracellular matrix synthesis and promoted the migration of microphage cells RAW264.7. CONCLUSIONS Plasma fractalkine levels were associated with creatinine level, 24-h urine protein, mesangial hypercellularity pathological damage, the CD68+ macrophage and CD20+ B cell infiltration in renal tissue and renal outcome in IgAN patients. Plasma fractalkine might be a potential prognosis novel predictor in Chinese patients with IgAN.
Collapse
Affiliation(s)
- Ran Luo
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shui-Ming Guo
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yue-Qiang Li
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Yang
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng-Lan Li
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Han
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Feng He
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shu-Wang Ge
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gang Xu
- Department of Nephrology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
75
|
Cormack S, Mohammed A, Panahi P, Das R, Steel AJ, Chadwick T, Bryant A, Egred M, Stellos K, Spyridopoulos I. Effect of ciclosporin on safety, lymphocyte kinetics and left ventricular remodelling in acute myocardial infarction. Br J Clin Pharmacol 2020; 86:1387-1397. [PMID: 32067256 PMCID: PMC7318996 DOI: 10.1111/bcp.14252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/12/2019] [Accepted: 12/23/2019] [Indexed: 11/27/2022] Open
Abstract
Aims Following a favourable pilot trial using a single bolus of ciclosporin, it has been unclear why 2 large studies (CYCLE and CIRCUS) failed to prevent reperfusion injury and reduce infarct size in STEMI (ST elevation myocardial infarction). The purpose of this study was to assess the effect of ciclosporin on myocardial injury, left ventricular remodelling and lymphocyte kinetics in patients with acute STEMI undergoing primary percutaneous coronary intervention. Methods In this double‐blind, single centre trial, we randomly assigned 52 acute STEMI patients with an onset of pain of <6 hours and blocked culprit artery to a single bolus of ciclosporin (n = 26) or placebo (n = 26, control group) prior to reperfusion by stent percutaneous coronary intervention. The primary endpoint was infarct size at 12 weeks. Results Mean infarct size at 12 weeks was identical in both groups (9.1% [standard deviation= 7.0] vs 9.1% [standard deviation = 7.0], P = .99; 95% confidence interval for difference: −4.0 to 4.1). CD3 T‐lymphocytes dropped to similar levels at 90 minutes (867 vs 852 cells/μL, control vs ciclosporin) and increased to 1454 vs 1650 cells/μL at 24 hours. Conclusion In our pilot trial, a single ciclosporin bolus did not affect infarct size or left ventricular remodelling, matching the results from CYCLE and CIRCUS. Our study suggests that ciclosporin does either not reach ischaemic cardiomyocytes, or requires earlier application during first medical contact. Finally, 1 bolus of ciclosporin is not sufficient to inhibit CD4 T‐lymphocyte proliferation during remodelling. We therefore believe that further studies are warranted. (Evaluating the effectiveness of intravenous Ciclosporin on reducing reperfusion injury in pAtients undergoing PRImary percutaneous coronary intervention [CAPRI]; NCT02390674)
Collapse
Affiliation(s)
- Suzanne Cormack
- Freeman Hospital, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | | | | | - Rajiv Das
- Freeman Hospital, Newcastle upon Tyne, UK.,Faculty of Health and Life Sciences, Northumbria University, UK
| | - Alison J Steel
- Newcastle Clinical Trials Unit, Faculty of Medical Sciences, Newcastle University, UK
| | - Thomas Chadwick
- Population Health Sciences Institute, Newcastle University, UK
| | - Andrew Bryant
- Population Health Sciences Institute, Newcastle University, UK
| | | | - Konstantinos Stellos
- Freeman Hospital, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Ioakim Spyridopoulos
- Freeman Hospital, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | | |
Collapse
|
76
|
Iking J, Klose J, Staniszewska M, Fendler WP, Herrmann K, Rischpler C. Imaging inflammation after myocardial infarction: implications for prognosis and therapeutic guidance. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:35-50. [PMID: 32077669 DOI: 10.23736/s1824-4785.20.03232-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation after myocardial infarction (MI) has been in the focus of cardiovascular research for several years as it influences the remodeling process of the ischemic heart and thereby critically determines the clinical outcome of the patient. Today, it is well appreciated that inflammation is a crucial necessity for the initiation of the natural wound healing process; however, excessive inflammation can have detrimental effects and might result in adverse ventricular remodeling which is associated with an increased risk of heart failure. Newly emerged imaging techniques facilitate the non-invasive assessment of immune cell infiltration into the ischemic myocardium and can provide greater insight into the underlying complex and dynamic repair mechanisms. Molecular imaging of inflammation in the context of MI may help with stratification of patients at high risk of adverse ventricular remodeling post-MI which may be of diagnostic, therapeutic, and prognostic value. Novel radiopharmaceuticals may additionally provide a way to combine patient monitoring and therapy. In spite of great advances in recent years in the field of imaging sciences, clinicians still need to overcome some obstacles to a wider implementation of inflammation imaging post-MI. This review focuses on inflammation as a molecular imaging target and its potential implication in prognosis and therapeutic guidance.
Collapse
Affiliation(s)
- Janette Iking
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.,Department of Cardiology I for Coronary and Peripheral Vascular Disease, and Heart Failure, University Hospital Münster, Münster, Germany
| | - Jasmin Klose
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
77
|
Ischemia/Reperfusion Injury: Pathophysiology, Current Clinical Management, and Potential Preventive Approaches. Mediators Inflamm 2020; 2020:8405370. [PMID: 32410868 PMCID: PMC7204323 DOI: 10.1155/2020/8405370] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/18/2019] [Accepted: 01/03/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial ischemia reperfusion syndrome is a complex entity where many inflammatory mediators play different roles, both to enhance myocardial infarction-derived damage and to heal injury. In such a setting, the establishment of an effective therapy to treat this condition has been elusive, perhaps because the experimental treatments have been conceived to block just one of the many pathogenic pathways of the disease, or because they thwart the tissue-repairing phase of the syndrome. Either way, we think that a discussion about the pathophysiology of the disease and the mechanisms of action of some drugs may shed some clarity on the topic.
Collapse
|
78
|
Chen T, Yang M. Platelet-to-lymphocyte ratio is associated with cardiovascular disease in continuous ambulatory peritoneal dialysis patients. Int Immunopharmacol 2019; 78:106063. [PMID: 31835088 DOI: 10.1016/j.intimp.2019.106063] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) patients have a high incidence of cardiovascular disease (CVD) events, which is related to the inflammatory status of this population. Platelet-to-lymphocyte ratio (PLR) is a relatively new indicator of inflammation. The aim of this study was to investigate the relationship between PLR and the CVD events in continuous ambulatory peritoneal dialysis (CAPD) patients. METHODS A total of 70 stable CAPD patients were included in this study from February 2014 to March 2017, and complete demographic characteristics and clinical laboratory baseline data were collected at enrollment. The primary endpoint was defined as the experienced of CVD events during the follow-up period. Binary logistic regression was used to assess the association between PLR and CVD events in CAPD patients. RESULTS During a median follow-up period of 22 months, 28 (40%) CAPD patients experienced CVD events. Patients in the CVD event group had a high level of platelets (P < 0.01), C-reactive protein (P < 0.01), PLR (P < 0.01) and neutrophil-to-lymphocyte ratio (NLR) (P < 0.01). However, lymphocyte counts (P < 0.01) were significantly lower than patients without CVD events. Following adjusted binary regression analysis revealed no relationship between high NLR and CVD events (OR, 1.21; 95% CI, 0.52-2.85; p = 0.44). However, the correlation between high PLR and CVD events was significant (OR, 1.05; 95% CI, 1.02-1.08; p < 0.01). High PLR was confirmed as an independent predictor of CVD events. CONCLUSION Our results demonstrated that PLR was independently associated with CVD events. High PLR can be used to predict the risk of CVD events in CAPD patients. PLR was easy to obtain and can be considered as a routine test to serve the clinic.
Collapse
Affiliation(s)
- Tianlei Chen
- Department of Nephrology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, PR China
| | - Min Yang
- Department of Nephrology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, PR China.
| |
Collapse
|
79
|
Rieckmann M, Delgobo M, Gaal C, Büchner L, Steinau P, Reshef D, Gil-Cruz C, Horst ENT, Kircher M, Reiter T, Heinze KG, Niessen HW, Krijnen PA, van der Laan AM, Piek JJ, Koch C, Wester HJ, Lapa C, Bauer WR, Ludewig B, Friedman N, Frantz S, Hofmann U, Ramos GC. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J Clin Invest 2019; 129:4922-4936. [PMID: 31408441 DOI: 10.1172/jci123859] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
T cell autoreactivity is a hallmark of autoimmune diseases but can also benefit self-maintenance and foster tissue repair. Herein, we investigated whether heart-specific T cells exert salutary or detrimental effects in the context of myocardial infarction (MI), the leading cause of death worldwide. After screening more than 150 class-II-restricted epitopes, we found that myosin heavy chain alpha (MYHCA) was a dominant cardiac antigen triggering post-MI CD4+ T cell activation in mice. Transferred MYHCA614-629-specific CD4+ T (TCR-M) cells selectively accumulated in the myocardium and mediastinal lymph nodes (med-LN) of infarcted mice, acquired a Treg phenotype with a distinct pro-healing gene expression profile, and mediated cardioprotection. Myocardial Treg cells were also detected in autopsies from patients who suffered a MI. Noninvasive PET/CT imaging using a CXCR4 radioligand revealed enlarged med-LNs with increased cellularity in MI-patients. Notably, the med-LN alterations observed in MI patients correlated with the infarct size and cardiac function. Taken together, the results obtained in our study provide evidence showing that MI-context induces pro-healing T cell autoimmunity in mice and confirms the existence of an analogous heart/med-LN/T cell axis in MI patients.
Collapse
Affiliation(s)
- Max Rieckmann
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Murilo Delgobo
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Chiara Gaal
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Lotte Büchner
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Philipp Steinau
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Dan Reshef
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Ellis N Ter Horst
- Heart Center, Amsterdam UMC, location AMC, Amsterdam, Netherlands.,Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Malte Kircher
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Theresa Reiter
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Hans Wm Niessen
- Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | - Paul Aj Krijnen
- Department of Pathology and Cardiac Surgery, Amsterdam UMC, location VUmc, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | | | - Jan J Piek
- Heart Center, Amsterdam UMC, location AMC, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | - Charlotte Koch
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University Munich, Munich, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Wolfgang R Bauer
- Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Stefan Frantz
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Ulrich Hofmann
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Gustavo Campos Ramos
- Department of Internal Medicine III, University Clinic Halle, Halle, Germany.,Department of Internal Medicine I, and.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
80
|
Li C, Zong W, Zhang M, Tu Y, Zhou Q, Ni M, Li Z, Liu H, Zhang J. Increased Ratio of Circulating T-Helper 1 to T-Helper 2 Cells and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction: A Prospective Observational Study. Med Sci Monit 2019; 25:6034-6042. [PMID: 31407674 PMCID: PMC6703085 DOI: 10.12659/msm.913891] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background This study aimed to determine the association between CD4-positive T-helper (Th) cell subsets, T-helper 1 (Th1) and T-helper 2 (Th2) in patients with acute myocardial infarction (AMI) and the severity of coronary artery disease (CAD) determined by coronary artery angiography. Material/Methods Three groups of patients with AMI who underwent coronary angiography and percutaneous coronary intervention (PCI) included patients with stable CAD (n=35), ST-segment elevation myocardial infarction (STEMI) (n=30), and non-STEMI (NSTEMI) (n=35), and controls (n=33). Measurement of high-sensitivity cardiac troponin T (hs-cTnT) was performed. The numbers of circulating CD4-positive Th1 and Th2 cells were measured using flow cytometry. Plasma levels of interferon-γ (IFN-γ) and interleukin-4 (IL-4) were measured using enzyme-linked immunosorbent assay (ELISA). Results An increase in the Th1 lymphocyte population was associated with more CAD, and an increased Th1/Th2 ratio was found in patients with NSTEMI and STEMI (controls 7.27±2.98; stable CAD 7.58±2.52; NSTEMI 16.62±2.74; and STEMI 22.32±7.35) (P<0.001). The proportion of Th1 cells and the Th1/Th2 ratio increased as the number of affected arteries, the degree of stenosis, and the lesion length increased. At a median follow-up of 18.2 months, patients with CAD and an increased Th1/Th2 ratio had a significant increase in adverse cardiac events compared with patients with a reduced Th1/Th2 ratio (log-rank, P=0.042). Conclusions An increased ratio of circulating Th1 to Th2 cells in patients with AMI was associated with the severity of CAD determined by angiography.
Collapse
Affiliation(s)
- Chang Li
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Wenxia Zong
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Ming Zhang
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Yanming Tu
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Qiyu Zhou
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Mingke Ni
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Zhiyong Li
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Haizhen Liu
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| | - Jingyi Zhang
- Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
81
|
Hao W, Li M, Zhang Y, Zhang C, Wang P. Severity of chronic obstructive pulmonary disease with 'exacerbator with emphysema phenotype' is associated with potential biomarkers. Postgrad Med J 2019; 96:28-32. [PMID: 31375557 DOI: 10.1136/postgradmedj-2019-136599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/02/2019] [Accepted: 07/24/2019] [Indexed: 11/04/2022]
Abstract
OBJECTIVE The present study was designed to investigate the biomarkers levels of fractalkine (FKN), neutrophil elastase (NE) and matrix metalloproteinase-12 (MMP-12) in chronic obstructive pulmonary disease (COPD) with 'exacerbator with emphysema phenotype' and to evaluate the associations between the biomarkers levels and the severity of disease by spirometric measurements. METHODS A total of 84 COPD patients and 49 healthy controls were enrolled in our study. ELISA were utilised to detect the FKN, MMP-12 and NE in serum from all subjects. RESULTS FKN (p<0.001), NE (p=0.039) and MMP-12 (p<0.001) in serum of COPD patients showed higher levels than that of healthy control subjects. Serum FKN (p<0.001), MMP-12 (p<0.001) and NE (p=0.043) levels were significantly higher in severe and very severe COPD patients than that in mild and moderate COPD patients. Circulating FKN, MMP-12 and NE expression levels were significantly elevated (p<0.001) in COPD smokers compared with COPD non-smokers. The smoke pack years were negatively correlated with FEV1%pred (r=-0.5036), FEV1/FVC ratio (r=-0.2847) (FEV, forced expiratory volume; FVC, forced vital capacity). Similarly, we observed a strong positive correlation between the smoke pack years and serum levels of FKN (r=0.4971), MMP-12 (r=0.4315) and NE (r=0.2754). FEV1%pred was strongly negatively correlated with cytokine levels of FKN (r=-0.4367), MMP-12 (r=-0.3295) and NE (r=-0.2684). Likewise, FEV1/FVC ratio was negatively correlated with mediators of inflammation levels of FKN (r=-0.3867), MMP-12 (r=-0.2941) and NE (r=-0.2153). CONCLUSION Serum FKN, MMP-12 and NE concentrations in COPD patients are directly associated with the severity of COPD with 'exacerbator with emphysema phenotype'. This finding suggests that FKN, MMP-12 and NE might play an important role in the pathophysiology of COPD.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Respiratory and Critical Care Medicine, Yan'an University Affiliated Hospital, Yan'an, China .,Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yunqing Zhang
- Department of Respiratory and Critical Care Medicine, Yan'an University Affiliated Hospital, Yan'an, China
| | - Cailian Zhang
- Department of Respiratory and Critical Care Medicine, Yan'an University Affiliated Hospital, Yan'an, China
| | - Ping Wang
- Department of Respiratory and Critical Care Medicine, Yan'an University Affiliated Hospital, Yan'an, China
| |
Collapse
|
82
|
Hong D, Choi KH, Song YB, Lee JM, Park TK, Yang JH, Hahn JY, Choi JH, Choi SH, Kim SM, Choe Y, Kim EK, Chang SA, Lee SC, Oh JK, Gwon HC. Prognostic implications of post-percutaneous coronary intervention neutrophil-to-lymphocyte ratio on infarct size and clinical outcomes in patients with acute myocardial infarction. Sci Rep 2019; 9:9646. [PMID: 31273274 PMCID: PMC6609693 DOI: 10.1038/s41598-019-46117-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/24/2019] [Indexed: 11/09/2022] Open
Abstract
This study evaluated the prognostic implications of post-percutaneous coronary intervention (PCI) neutrophil-to-lymphocyte ratio (NLR) in patients with acute myocardial infarction (AMI). A total of 309 patients with AMI who underwent cardiac magnetic resonance imaging (CMR) and a complete blood cell count within 24 hours before and after PCI were enrolled. Primary outcome was infarct size. Patients were assigned to high (n = 118) or low (n = 191) NLR groups according to the best cut-off value of 3.88. Infarct size (% of total left ventricular mass) was significantly higher in the high NLR group than in the low NLR group (24.1 ± 11.0 vs. 16.7 ± 9.1, p < 0.001). Post-PCI NLR ≥ 3.88 was associated with risk of a large-sized infarction (≥20%) (OR 2.91, 95% CI 1.73-4.88, p < 0.001). The risk of MACE was also significantly higher in the high NLR group than in the low NLR group (15.8% vs. 7.4%, HR 2.60, 95% CI 1.21-5.60, p = 0.015). Among patients with AMI who underwent PCI, high post-PCI NLR value was associated with higher risk of large-sized infarction as measured by CMR, as well as adverse clinical outcomes. Our findings suggest that post-PCI NLR is a useful tool for risk assessment in patients with AMI who undergo PCI.
Collapse
Affiliation(s)
- David Hong
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Hong Choi
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Bin Song
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Joo Myung Lee
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taek Kyu Park
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Yang
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joo-Yong Hahn
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin-Ho Choi
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Emergency Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung-Hyuk Choi
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Mok Kim
- Department of Radiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yeonhyeon Choe
- Department of Radiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Kyoung Kim
- Cardiovascular Imaging Center, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung A Chang
- Cardiovascular Imaging Center, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Chol Lee
- Cardiovascular Imaging Center, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jae K Oh
- Cardiovascular Imaging Center, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hyeon-Cheol Gwon
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
83
|
Cheng Y, Zhao H, Song P, Zhang Z, Chen J, Zhou YH. Dynamic changes of lymphocyte counts in adult patients with severe pandemic H1N1 influenza A. J Infect Public Health 2019; 12:878-883. [PMID: 31202719 PMCID: PMC7102863 DOI: 10.1016/j.jiph.2019.05.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/15/2019] [Accepted: 05/27/2019] [Indexed: 11/24/2022] Open
Abstract
Background Lymphopenia has been observed in severe pandemic influenza A/H1N1 in developed countries. However, data from developing countries are rare and dynamic change of lymphocyte counts in severe pandemic influenza A/H1N1 is scarcely reported. This study aimed to observe change of lymphocyte counts in patients with severe pandemic influenza A/H1N1 and to investigate the correlation of lymphopenia and severe pandemic influenza A/H1N1. Methods We retrospectively analyzed the white blood cell counts and differentials and other clinical data in 21 hospitalized patients with severe pandemic influenza A/H1N1 confirmed by reverse-transcription PCR during 2009 and 2010. Results All patients, except two cases with bacterial co-infections, had normal or reduced white blood cell counts. Seventeen (81.0%) patients had decreased lymphocyte proportions (<20%) and counts (<0.8 × 109/L), with the lowest value of 1.2% and 0.1 × 109/L respectively. A patient with nosocomial infection of influenza A/H1N1 showed that lymphopenia occurred on the first day of illness. Lymphocyte proportions and absolute counts returned to normal or slightly higher than normal in 16 of the 17 patients within 2–3 weeks after the disease onset. Conclusions Lymphopenia along with other clinical parameters may be helpful in early differential diagnosis of severe pandemic influenza A/H1N1.
Collapse
Affiliation(s)
- Yandong Cheng
- Department of Internal Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Hong Zhao
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing 210003, China
| | - Peixin Song
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Zhaoping Zhang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Junhao Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China.
| | - Yi-Hua Zhou
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China; Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.
| |
Collapse
|
84
|
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
|
85
|
Wang J, Duan Y, Sluijter JPG, Xiao J. Lymphocytic subsets play distinct roles in heart diseases. Am J Cancer Res 2019; 9:4030-4046. [PMID: 31281530 PMCID: PMC6592175 DOI: 10.7150/thno.33112] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Heart diseases are one of the leading causes of death for humans in the world. Increasing evidence has shown that myocardial injury induced innate and adaptive immune responses upon early cellular damage but also during chronic phases post-injury. The immune cells can not only aggravate the injury but also play an essential role in the induction of wound healing responses, which means they play a complex role throughout the acute inflammatory response and reparative response after cardiac injury. This review will summarize the current experimental and clinical evidence of lymphocytes, one of the major types of immune cells, participate in heart diseases and try to explain the possible role of these immune cells following cardiac injury.
Collapse
|
86
|
Novel Molecular Targets Participating in Myocardial Ischemia-Reperfusion Injury and Cardioprotection. Cardiol Res Pract 2019; 2019:6935147. [PMID: 31275641 PMCID: PMC6558612 DOI: 10.1155/2019/6935147] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Worldwide morbidity and mortality from acute myocardial infarction (AMI) and related heart failure remain high. While effective early reperfusion of the criminal coronary artery after a confirmed AMI is the typical treatment at present, collateral myocardial ischemia-reperfusion injury (MIRI) and pertinent cardioprotection are still challenging to address and have inadequately understood mechanisms. Therefore, unveiling the related novel molecular targets and networks participating in triggering and resisting the pathobiology of MIRI is a promising and valuable frontier. The present study specifically focuses on the recent MIRI advances that are supported by sophisticated bio-methodology in order to bring the poorly understood interrelationship among pro- and anti-MIRI participant molecules up to date, as well as to identify findings that may facilitate the further investigation of novel targets.
Collapse
|
87
|
Neupane R, Jin X, Sasaki T, Li X, Murohara T, Cheng XW. Immune Disorder in Atherosclerotic Cardiovascular Disease - Clinical Implications of Using Circulating T-Cell Subsets as Biomarkers. Circ J 2019; 83:1431-1438. [PMID: 31092769 DOI: 10.1253/circj.cj-19-0114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerotic cardiovascular disease (ACVD) is an inflammatory phenomenon that leads to structural abnormality in the vascular lumen due to the formation of atheroma by the deposition of lipid particles and inflammatory cytokines. There is a close interaction between innate immune cells (neutrophils, monocyte, macrophages, dendritic cells) and adaptive immune cells (T and B lymphocytes) in the initiation and progression of atherosclerosis. According to novel insights into the role of adaptive immunity in atherosclerosis, the activation of CD4+T cells in response to oxidized low-density lipoprotein-antigen initiates the formation and facilitates the propagation of atheroma, whereas CD8+T cells cause the rupture of a developed atheroma by their cytotoxic nature. Peripheral CD4+and CD8+T-cell counts were altered in patients with other cardiovascular risk factors. Furthermore, on evaluation of the feasibility of immune cells as a diagnostic tool, the blood CD4+(helper), CD8+(cytotoxic), and CD4+CD25+Foxp3+(regulatory) T cells and the ratio of CD4 to CD8 cells hold promise as biomarkers of coronary artery disease and their subtypes. T cells also could be a therapeutic target for cardiovascular diseases. The goal of this review was therefore to summarize the available information regarding immune disorders in ACVD with a special focus on the clinical implications of circulating T-cell subsets as biomarkers.
Collapse
Affiliation(s)
- Rajib Neupane
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Xiongjie Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine
| | - Xiang Li
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital.,Department of Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|
88
|
Ji CL, Nomi A, Li B, Shen C, Song BC, Zhang JG. Increased Plasma Soluble Fractalkine in Patients with Chronic Heart Failure and Its Clinical Significance. Int Heart J 2019; 60:701-707. [PMID: 31019174 DOI: 10.1536/ihj.18-422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Fractalkine has been reported to play an important role in the pathophysiology of various cardiovascular disorders. This research aims to study the change of soluble fractalkine (sFKN) in plasma level of patients with chronic heart failure (CHF) and evaluate its prognostic value.A total of 96 patients with CHF and 45 healthy subjects were included in this study. The plasma levels of sFKN, brain natriuretic peptide (BNP), and Interleukin-18 (IL-18) were determined by ELISA kits when they were first admitted to the hospital. Left ventricular ejection fraction (LVEF) was measured by echocardiogram. Rehospitalization status within 1 year after the first hospitalization was also recorded.The plasma levels of sFKN, BNP, and IL-18 in patients with CHF were significantly higher than in the control group (P < 0.05). The concentrations of sFKN and BNP were increased with the severity of heart failure classified by NYHA classification (P < 0.05). There were no statistical differences among all CHF subgroups classified by etiology (P > 0.05). Plasma sFKN level in CHF group was positively correlated with BNP (r = 0.441, P < 0.001) and IL-18 (r = 0.592, P < 0.001). Receiver operating characteristic curve analysis showed that area under the curve values of FKN, BNP, and IL-18 were 0.885 (95%CI: 0.810 to 0.960, P < 0.001), 0.889 (95%CI: 0.842 to 0.956, P < 0.001), and 0.878 (95%CI: 0.801-0.954, P < 0.001), respectively. The concentrations of sFKN and BNP were increased in patients readmitted more than once within 1 year (P < 0.05).
Collapse
Affiliation(s)
- Cui-Ling Ji
- Department of Cardiology II, The Affiliated Hospital of Jining Medical University
| | - Adnan Nomi
- Teaching and Research Section of International Students, Jining Medical University
| | - Bin Li
- Department of Cardiology IV, The Affiliated Hospital of Jining Medical University
| | - Cheng Shen
- Department of Cardiology II, The Affiliated Hospital of Jining Medical University
| | - Bing-Chun Song
- Department of Cardiology II, The Affiliated Hospital of Jining Medical University
| | - Jin-Guo Zhang
- Department of Cardiology II, The Affiliated Hospital of Jining Medical University
| |
Collapse
|
89
|
Growth Factor Screening in Dystrophic Muscles Reveals PDGFB/PDGFRB-Mediated Migration of Interstitial Stem Cells. Int J Mol Sci 2019; 20:ijms20051118. [PMID: 30841538 PMCID: PMC6429448 DOI: 10.3390/ijms20051118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 01/08/2023] Open
Abstract
Progressive muscle degeneration followed by dilated cardiomyopathy is a hallmark of muscular dystrophy. Stem cell therapy is suggested to replace diseased myofibers by healthy myofibers, although so far, we are faced by low efficiencies of migration and engraftment of stem cells. Chemokines are signalling proteins guiding cell migration and have been shown to tightly regulate muscle tissue repair. We sought to determine which chemokines are expressed in dystrophic muscles undergoing tissue remodelling. Therefore, we analysed the expression of chemokines and chemokine receptors in skeletal and cardiac muscles from Sarcoglycan-α null, Sarcoglycan-β null and immunodeficient Sgcβ-null mice. We found that several chemokines are dysregulated in dystrophic muscles. We further show that one of these, platelet-derived growth factor-B, promotes interstitial stem cell migration. This finding provides perspective to an approachable mechanism for improving stem cell homing towards dystrophic muscles.
Collapse
|
90
|
Santos-Zas I, Lemarié J, Tedgui A, Ait-Oufella H. Adaptive Immune Responses Contribute to Post-ischemic Cardiac Remodeling. Front Cardiovasc Med 2019; 5:198. [PMID: 30687720 PMCID: PMC6335242 DOI: 10.3389/fcvm.2018.00198] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/21/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is a common condition responsible for mortality and morbidity related to ischemic heart failure. Accumulating experimental and translational evidence support a crucial role for innate immunity in heart failure and adverse heart remodeling following MI. More recently, the role of adaptive immunity in myocardial ischemia has been identified, mainly in rodents models of both transient and permanent heart ischemia. The present review summarizes the experimental evidence regarding the role of lymphocytes and dendritic cells in myocardial remodeling following coronary artery occlusion. Th1 and potentially Th17 CD4+ T cell responses promote adverse heart remodeling, whereas regulatory T cells appear to be protective, modulating macrophage activity, cardiomyocyte survival, and fibroblast phenotype. The role of CD8+ T cells in this setting remains unknown. B cells contribute to adverse cardiac remodeling through the modulation of monocyte trafficking, and potentially the production of tissue-specific antibodies. Yet, further substantial efforts are still required to confirm experimental data in human MI before developing new therapeutic strategies targeting the adaptive immune system in ischemic cardiac diseases.
Collapse
Affiliation(s)
- Icia Santos-Zas
- INSERM UMR-S 970, Sorbonne Paris Cité, Paris Cardiovascular Research Center - PARCC, Université Paris Descartes, Paris, France
| | - Jérémie Lemarié
- INSERM UMR-S 970, Sorbonne Paris Cité, Paris Cardiovascular Research Center - PARCC, Université Paris Descartes, Paris, France.,UMR_S 1116, Université de Lorraine, Inserm, DCAC, Centre Hospitalier Régional Universitaire de Nancy - Réanimation Médicale - Hôpital Central, Nancy, France
| | - Alain Tedgui
- INSERM UMR-S 970, Sorbonne Paris Cité, Paris Cardiovascular Research Center - PARCC, Université Paris Descartes, Paris, France
| | - Hafid Ait-Oufella
- INSERM UMR-S 970, Sorbonne Paris Cité, Paris Cardiovascular Research Center - PARCC, Université Paris Descartes, Paris, France.,AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Saint-Antoine, Sorbonne Université, Paris, France
| |
Collapse
|
91
|
Diagnostic Value of Parameters Related to White Blood Cell Counts for Troponin I Elevation in CO Poisoning. Cardiovasc Toxicol 2019; 19:334-343. [DOI: 10.1007/s12012-018-09501-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
92
|
Wang A, Yang T, Zhang L, Jia L, Wu Q, Yao S, Xu J, Yang H. IP3-Mediated Calcium Signaling Is Involved in the Mechanism of Fractalkine-Induced Hyperalgesia Response. Med Sci Monit 2018; 24:8804-8811. [PMID: 30517088 PMCID: PMC6290586 DOI: 10.12659/msm.913787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Fractalkine is widely expressed throughout the brain and spinal cord, where it can exert effects on pain enhancement and hyperalgesia by activating microglia through CX3C chemokine receptor 1 (CX3CR1), which triggers the release of several pro-inflammatory cytokines in the spinal cord. Fractalkine has also been shown to increase cytosolic calcium ([Ca2+]i) in microglia. Material/Methods Based on the characteristics of CX3CR1, a G protein-coupled receptor, we explored the role of inositol 1,4,5-trisphosphate (IP3) signaling in fractalkine-induced inflammatory response in BV-2 cells in vitro. The effect and the underlying mechanism induced by fractalkine in the brain were observed using a mouse model with intracerebroventricular (i.c.v.) injection of exogenous fractalkine. Results [Ca2+]i was significantly increased and IL-1β and TNF-α levels were higher in the fractalkine-treated cell groups than in the farctalkine+ 2-APB groups. We found that i.c.v. injection of fractalkine significantly increased p-p38MAPK, IL-1β, and TNF-α expression in the brain, while i.c.v. injection of a fractalkine-neutralizing antibody (anti-CX3CR1), trisphosphate receptor (IP3R) antagonist (2-APB), or p38MAPK inhibitor (SB203580) prior to fractalkine addition yielded an effective and reliable anti-allodynia effect, following the reduction of p-p38MAPK, IL-1β, and TNF-α expression. Conclusions Our results suggest that fractalkine leads to hyperalgesia, and the underlying mechanism may be associated with IP3/p38MAPK-mediated calcium signaling and its phlogogenic properties.
Collapse
Affiliation(s)
- Aitao Wang
- Department of Anesthesiology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China (mainland)
| | - Tingting Yang
- Key Laboratory of Antibody Technique of National Health and Family Planning Commission, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Lingli Zhang
- Department of Ophthalmology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China (mainland)
| | - Lizhou Jia
- Key Laboratory of Antibody Technique of National Health and Family Planning Commission, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jianjun Xu
- Department of Anesthesiology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang, China (mainland)
| | - Hongxin Yang
- Department of Pharmacy, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China (mainland)
| |
Collapse
|
93
|
Owens AP, Robbins N, Saum K, Jones SM, Kirschner A, Woo JG, McCoy C, Slone S, Rothenberg ME, Urbina EM, Tranter M, Rubinstein J. Tefillin use induces remote ischemic preconditioning pathways in healthy men. Am J Physiol Heart Circ Physiol 2018; 315:H1748-H1758. [PMID: 30216115 DOI: 10.1152/ajpheart.00347.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The present study assessed whether tefillin use (tight, nonocclusive, wrapping of the arm) elicits a remote ischemic preconditioning (RIPC)-like effect in subjects with both acute and chronic use. RIPC, created by short bursts of ischemia-reperfusion, has not been successfully taken to the bedside. Several large population studies have found that Orthodox Jewish men (who wear tefillin almost daily) have decreased cardiovascular mortality compared with non-Orthodox counterparts. We hypothesized that tefillin use is a relevant component in triggering a preconditioning effect. Jewish men ( n = 20) were enrolled; 9 men were daily tefillin users (conditioned) and 11 men were nonusers of tefillin as controls (naïve). Subjects were evaluated for adherence to traditional Jewish practice, had vital signs measured, blood drawn for analysis of circulating cytokines and monocyte function, and underwent brachial flow-mediated dilation to evaluate vascular reactivity at baseline (basal) and after 30 min of using tefillin (acute treatment). Under basal conditions, both groups had similar peak systolic velocity (SV), diameter, and flow volume, although the conditioned group had higher SV at 120 s postdeflation ( P = 0.05). Acute tefillin use augmented artery diameter and flow volume in both groups, with conditioned subjects experiencing higher SV than control subjects at 90 and 120 s postdeflation ( P = 0.03 and P = 0.02, respectively). Conditioned subjects had decreased inflammation, monocyte migration and adhesion, and endothelial activation compared with control subjects at baseline. Acute use of tefillin did not significantly alter monocyte function in either group. In this pilot study, acute tefillin use improves vascular function, whereas chronic tefillin use is associated with an anti-inflammatory RIPC-like phenotype. NEW & NOTEWORTHY We hypothesized that tefillin use among Orthodox Jewish men (who practice a nonocclusive leather banding of their nondominant arm) will induce a remote ischemic preconditioning phenotype. Chronic use of tefillin in Orthodox Jewish men was associated with increased systolic velocity and attenuated inflammation and monocyte chemotaxis and adhesion versus Jewish men who do not wear tefillin. Acute use of tefillin in both populations augmented brachial artery diameter and blood flow but not inflammatory profiles compared with baseline.
Collapse
Affiliation(s)
- A Phillip Owens
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Nathan Robbins
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Keith Saum
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio.,Department of Biomedical Engineering, University of Cincinnati , Cincinnati, Ohio
| | - Shannon M Jones
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Akiva Kirschner
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Jessica G Woo
- Department of Pediatrics, University of Cincinnati , Cincinnati, Ohio.,Division of Biostatistics and Epidemiology, University of Cincinnati , Cincinnati, Ohio
| | - Connie McCoy
- Department of Pediatrics, University of Cincinnati , Cincinnati, Ohio.,Division of Cardiology, University of Cincinnati , Cincinnati, Ohio
| | - Samuel Slone
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Marc E Rothenberg
- Department of Pediatrics, University of Cincinnati , Cincinnati, Ohio.,Division of Allergy and Immunology, University of Cincinnati , Cincinnati, Ohio
| | - Elaine M Urbina
- Department of Pediatrics, University of Cincinnati , Cincinnati, Ohio.,Division of Cardiology, University of Cincinnati , Cincinnati, Ohio
| | - Michael Tranter
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Jack Rubinstein
- Division of Cardiovascular Health and Disease, Department of Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
94
|
Xu B, Qian Y, Zhao Y, Fang Z, Tang K, Zhou N, Li D, Wang J. Prognostic value of fractalkine/CX3CL1 concentration in patients with acute myocardial infarction treated with primary percutaneous coronary intervention. Cytokine 2018; 113:365-370. [PMID: 30352758 DOI: 10.1016/j.cyto.2018.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/25/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent studies demonstrated that fractalkine (FKN) is critically involved in the regulation of inflammation and cardiac function. OBJECTIVE This study aimed to investigate the prognostic value of circulating FKN in patients with ST-elevated acute myocardial infarction (STEMI) after primary PCI. METHODS We enrolled ninety consecutive STEMI patients and investigated the association of circulating FKN with myocardial salvage and the occurrence of major adverse cardiac events (MACE) after PCI. RESULTS During a median follow-up of 387 days, total 15 MACE (16.67%) were registered in the study population. Patients with MACE were more likely to be occurred in elderly patients with 3-vessel disease. Correlation analysis demonstrated the level of FKN at day 1 after PCI (FKN@day-1) not only significantly correlated with the levels of hs-TnT at day 7 after PCI (R2 = 0.06; p = 0.02) but inversely correlated with the measurements of LVEF at 1-month observation (R2 = 0.10; p = 0.00). Kaplan-Meier survival analyses further revealed that patients with the level of FKN@day-1 above the median had a higher incidence of MACE compared with those whose FKN@day-1 levels below the median (log-rank test x2 = 13.29, p < 0.001). In addition, multivariate Cox regression analysis demonstrated that FKN@day-1 was an independent predictor of MACE (hazard ratio: 4.63; 95% confidence interval: 1.53-14.01; p = 0.00), together with WBC count and 3-vessel disease for STEMI patients. CONCLUSIONS Our study demonstrates that FKN@day-1 is negative correlated with myocardial salvage after acute myocardial infarction and might be a valuable prognostic marker of MACE in patients with STEMI undergone PCI.
Collapse
Affiliation(s)
- Bing Xu
- Department of Cardiology, Northern Jiangsu Province Hospital and Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yanxia Qian
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yingming Zhao
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zhen Fang
- Department of Cardiology, Northern Jiangsu Province Hospital and Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Kangting Tang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ningtian Zhou
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Dianfu Li
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Junhong Wang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China; Department of Cardiology, The People's Hospital of Kizilsu Kirghiz Autonomous Prefecture, Xinjiang, China.
| |
Collapse
|
95
|
FPR1 gene silencing suppresses cardiomyocyte apoptosis and ventricular remodeling in rats with ischemia/reperfusion injury through the inhibition of MAPK signaling pathway. Exp Cell Res 2018; 370:506-518. [DOI: 10.1016/j.yexcr.2018.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/25/2022]
|
96
|
Huang S, Frangogiannis NG. Anti-inflammatory therapies in myocardial infarction: failures, hopes and challenges. Br J Pharmacol 2018; 175:1377-1400. [PMID: 29394499 PMCID: PMC5901181 DOI: 10.1111/bph.14155] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/14/2022] Open
Abstract
In the infarcted heart, the damage-associated molecular pattern proteins released by necrotic cells trigger both myocardial and systemic inflammatory responses. Induction of chemokines and cytokines and up-regulation of endothelial adhesion molecules mediate leukocyte recruitment in the infarcted myocardium. Inflammatory cells clear the infarct of dead cells and matrix debris and activate repair by myofibroblasts and vascular cells, but may also contribute to adverse fibrotic remodelling of viable segments, accentuate cardiomyocyte apoptosis and exert arrhythmogenic actions. Excessive, prolonged and dysregulated inflammation has been implicated in the pathogenesis of complications and may be involved in the development of heart failure following infarction. Studies in animal models of myocardial infarction (MI) have suggested the effectiveness of pharmacological interventions targeting the inflammatory response. This article provides a brief overview of the cell biology of the post-infarction inflammatory response and discusses the use of pharmacological interventions targeting inflammation following infarction. Therapy with broad anti-inflammatory and immunomodulatory agents may also inhibit important repair pathways, thus exerting detrimental actions in patients with MI. Extensive experimental evidence suggests that targeting specific inflammatory signals, such as the complement cascade, chemokines, cytokines, proteases, selectins and leukocyte integrins, may hold promise. However, clinical translation has proved challenging. Targeting IL-1 may benefit patients with exaggerated post-MI inflammatory responses following infarction, not only by attenuating adverse remodelling but also by stabilizing the atherosclerotic plaque and by inhibiting arrhythmia generation. Identification of the therapeutic window for specific interventions and pathophysiological stratification of MI patients using inflammatory biomarkers and imaging strategies are critical for optimal therapeutic design.
Collapse
Affiliation(s)
- Shuaibo Huang
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology)Albert Einstein College of MedicineBronxNY10461USA
- Department of Cardiology, Changzheng HospitalSecond Military Medical UniversityShanghai200003China
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology)Albert Einstein College of MedicineBronxNY10461USA
| |
Collapse
|
97
|
Impact of stress on aged immune system compartments: Overview from fundamental to clinical data. Exp Gerontol 2018; 105:19-26. [DOI: 10.1016/j.exger.2018.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 12/12/2022]
|
98
|
Apoptosis and Mobilization of Lymphocytes to Cardiac Tissue Is Associated with Myocardial Infarction in a Reperfused Porcine Model and Infarct Size in Post-PCI Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1975167. [PMID: 29743973 PMCID: PMC5878889 DOI: 10.1155/2018/1975167] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023]
Abstract
ST-segment elevation myocardial infarction (STEMI) is the most severe outcome of coronary artery disease. Despite rapid reperfusion of the artery, acute irrigation of the cardiac tissue is associated with increased inflammation. While innate immune response in STEMI is well described, an in-depth characterization of adaptive immune cell dynamics and their potential role remains elusive. We performed a translational study using a controlled porcine reperfusion model of STEMI and the analysis of lymphocyte subsets in 116 STEMI patients undergoing percutaneous coronary intervention (PCI). In the animal model, a sharp drop in circulating T lymphocytes occurred within the first hours after reperfusion. Notably, increased apoptosis of circulating lymphocytes and infiltration of proinflammatory Th1 lymphocytes in the heart were observed 48 h after reperfusion. Similarly, in STEMI patients, a sharp drop in circulating T lymphocyte subsets occurred within the first 24 h post-PCI. A cardiac magnetic resonance (CMR) evaluation of these patients revealed an inverse association between 24 h circulating T lymphocyte numbers and infarction size at 1-week and 6-month post-PCI. Our translational approach revealed striking changes in the circulating and tissue-infiltrating T lymphocyte repertoire in response to ischemia-reperfusion. These findings may help in developing new diagnostic and therapeutic approaches for coronary diseases.
Collapse
|
99
|
Horckmans M, Bianchini M, Santovito D, Megens RT, Springael JY, Negri I, Vacca M, Di Eusanio M, Moschetta A, Weber C, Duchene J, Steffens S. Pericardial Adipose Tissue Regulates Granulopoiesis, Fibrosis, and Cardiac Function After Myocardial Infarction. Circulation 2018; 137:948-960. [DOI: 10.1161/circulationaha.117.028833] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022]
Abstract
Background:
The pericardial adipose tissue (AT) contains a high density of lymphoid clusters. It is unknown whether these clusters play a role in post–myocardial infarction (MI) inflammatory responses and cardiac outcome.
Methods:
Lymphoid clusters were examined in epicardial AT of humans with or without coronary artery disease. Murine pericardial lymphoid clusters were visualized in mice subjected to coronary artery ligation. To study the relevance of pericardial clusters during inflammatory responses after MI, we surgically removed the pericardial AT and performed B-cell depletion and granulocyte-macrophage colony-stimulating factor blockade. Leukocytes in murine hearts, pericardial AT, spleen, mediastinal lymph nodes, and bone marrow were quantified by flow cytometry. Cannabinoid receptor CB2 (CB2
–/–
) mice were used as a model for enhanced B-cell responses. The effect of impaired dendritic cell (DC) trafficking on pericardial AT inflammatory responses was tested in CCR7
–/–
mice subjected to MI. Cardiac fibrosis and ventricular function were assessed by histology and echocardiography.
Results:
We identified larger B-cell clusters in epicardial AT of human patients with coronary artery disease in comparison with controls without coronary artery disease. Infarcted mice also had larger pericardial clusters and 3-fold upregulated numbers of granulocyte-macrophage colony-stimulating factor–producing B cells within pericardial AT, but not spleen or lymph nodes. This was associated with higher DC and T-cell counts in pericardial AT, which outnumbered DCs and T cells in lymph nodes. Analysis of DC maturation markers, tracking experiments with fluorescently labeled cells, and use of CCR7-deficient mice suggested that activated DCs migrate from infarcts into pericardial AT via CCR7. B-cell depletion or granulocyte-macrophage colony-stimulating factor neutralization inhibited DC and T-cell expansion within pericardial AT, and translated into reduced bone marrow granulopoiesis and cardiac neutrophil infiltration 3 days after MI. The relevance of the pericardial AT in mediating all these effects was confirmed by removal of pericardial AT and ex vivo coculture with pericardial AT and granulocyte progenitors. Finally, enhanced fibrosis and worsened ejection fraction in CB2
–/–
mice were limited by pericardial AT removal.
Conclusions:
Our findings unveil a new mechanism by which the pericardial AT coordinates immune cell activation, granulopoiesis, and outcome after MI.
Collapse
Affiliation(s)
| | - Mariaelvy Bianchini
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
| | - Remco T.A. Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
- Department of Biomedical Engineering (R.T.A.M.)
| | | | | | - Michele Vacca
- Cardiovascular Research Institute Maastricht, Maastricht University, Netherlands. Department of Interdisciplinary Medicine (M.V.)
- University of Bari “Aldo Moro,” Italy. Ageing Research Center, “G. d’Annunzio” University Foundation, Chieti, Italy (M.V.)
- University of Cambridge, United Kingdom (M.V.)
| | - Marco Di Eusanio
- Cardiovascular Department, “S.Orsola Malpighi” Hospital, University of Bologna, Italy (M.D.E.)
- Cardiac Surgery Unit, Ospedali Riuniti of Ancona, Italy (M.D.E.)
- Cardiac Surgery Department, Ospedali Riuniti “Umberto I-Lancisi-Salesi,” Ancona, Italy (M.D.E.)
| | | | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
- Department of Biochemistry (C.W.)
- German Center for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C.W., S.S.)
| | - Johan Duchene
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.B., D.S., R.T.A.M., C.W., J.D., S.S.)
- German Center for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C.W., S.S.)
| |
Collapse
|
100
|
Chen B, Frangogiannis NG. Immune cells in repair of the infarcted myocardium. Microcirculation 2018; 24. [PMID: 27542099 DOI: 10.1111/micc.12305] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022]
Abstract
The immune system plays a critical role in both repair and remodeling of the infarcted myocardium. Danger signals released by dying cardiomyocytes mobilize, recruit, and activate immune cells, triggering an inflammatory reaction. CXC chemokines containing the ELR motif attract neutrophils, while CC chemokines mediate recruitment of mononuclear cell subpopulations, contributing to clearance of the infarct from dead cells and matrix debris. Immune cell subsets also participate in suppression and containment of the postinfarction inflammatory response by secreting anti-inflammatory mediators, such as IL-10 and TGF-β. As proinflammatory signaling is suppressed, macrophage subpopulations, mast cells and lymphocytes, activate fibrogenic and angiogenic responses, contributing to scar formation. In the viable remodeling myocardium, chronic activation of immune cells may promote fibrosis and hypertrophy. This review discusses the role of immune cells in repair and remodeling of the infarcted myocardium. Understanding the role of immune cells in myocardial infarction is critical for the development of therapeutic strategies aimed at protecting the infarcted heart from adverse remodeling. Moreover, modulation of immune cell phenotype may be required in order to achieve the visionary goal of myocardial regeneration.
Collapse
Affiliation(s)
- Bijun Chen
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|